1
|
Zheng X, Huang H, Zhou Z, Guo W, Yang G, Chen Z, Chen D, Chen Y, Yuan G. Axin1 regulates tooth root development by inhibiting AKT1-mTORC1 activation and Shh translation in Hertwig's epithelial root sheath. Development 2024; 151:dev202899. [PMID: 39344774 DOI: 10.1242/dev.202899] [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: 03/26/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Hertwig's epithelial root sheath (HERS) interacts with dental apical mesenchyme and guides development of the tooth root, which is integral to the function of the whole tooth. However, the key genes in HERS essential for root development are understudied. Here, we show that Axin1, a scaffold protein that negatively regulates canonical Wnt signaling, is strongly expressed in the HERS. Axin1 ablation in the HERS of mice leads to defective root development, but in a manner independent of canonical Wnt signaling. Further studies reveal that Axin1 in the HERS negatively regulates the AKT1-mTORC1 pathway through binding to AKT1, leading to inhibition of ribosomal biogenesis and mRNA translation. Sonic hedgehog (Shh) protein, a morphogen essential for root development, is over-synthesized by upregulated mTORC1 activity upon Axin1 inactivation. Importantly, either haploinsufficiency of the mTORC1 subunit Rptor or pharmacological inhibition of Shh signaling can rescue the root defects in Axin1 mutant mice. Collectively, our data suggest that, independently of canonical Wnt signaling, Axin1 controls ribosomal biogenesis and selective mRNA translation programs via AKT1-mTORC1 signaling during tooth root development.
Collapse
Affiliation(s)
- Xiaoyu Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Hongcan Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Zhipeng Zhou
- National Key Laboratory of Agricultural Microbiology, College of Life Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Weihua Guo
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, Yunnan 610041, China
- Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan 610041, China
| | - Guobin Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Zhi Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institutes of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Guohua Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| |
Collapse
|
2
|
Lu C, Lv Y, Meng X, Yang T, Liu Y, Kou G, Yang X, Luo J. The potential toxic effects of estrogen exposure on neural and vascular development in zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116862. [PMID: 39128450 DOI: 10.1016/j.ecoenv.2024.116862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Estrogens and estrogenic chemicals are endocrine-disrupting chemicals (EDCs). The potential toxicity of EDCs to humans and aquatic organisms has become increasingly concerning. However, at present, the potential toxic mechanisms of EDCs on neural and vascular development are still being fully investigated. During the study, we utilized zebrafish to assess the developmental neural and vascular toxicity of different estrogens. The results indicated that zebrafish treated with different estrogens, especially E2, exhibit developmental malformations, including increased mortality, decreased body length, decreased heart rate, aberrant swimming behavior, and increased developmental malformations, including spinal curvature (SC), yolk edema (YE) and pericaidial edema (PE), in a dose-dependent manner with 72 h-treated. Further morphological evaluation revealed that E2 exposure significantly induced motor neural abnormalities in zebrafish embryos. In addition, treated with these three estrogens also impaired the vascular development in the early stage of zebrafish embryos. Mechanistically, the identification of downstream factors revealed that several key neural and vascular development-related genes, including syn2a, gfap, gap43, shha, kdr, flt1 and flt4, were transcriptionally downregulated after estrogen exposure in zebrafish, suggesting that estrogen exposure might cause neural and vascular toxicity by interfering the mRNA levels of genes relevant to neural and vascular development.
Collapse
Affiliation(s)
- Chunjiao Lu
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou 515041, China
| | - Yuhang Lv
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou 515041, China
| | - Xin Meng
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou 515041, China
| | - Ting Yang
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou 515041, China
| | - Yi Liu
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou 515041, China
| | - Guanhua Kou
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou 515041, China
| | - Xiaojun Yang
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou 515041, China; Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China.
| | - Juanjuan Luo
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
3
|
Shalabi S, Belayachi A, Larrivée B. Involvement of neuronal factors in tumor angiogenesis and the shaping of the cancer microenvironment. Front Immunol 2024; 15:1284629. [PMID: 38375479 PMCID: PMC10875004 DOI: 10.3389/fimmu.2024.1284629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/09/2024] [Indexed: 02/21/2024] Open
Abstract
Emerging evidence suggests that nerves within the tumor microenvironment play a crucial role in regulating angiogenesis. Neurotransmitters and neuropeptides released by nerves can interact with nearby blood vessels and tumor cells, influencing their behavior and modulating the angiogenic response. Moreover, nerve-derived signals may activate signaling pathways that enhance the production of pro-angiogenic factors within the tumor microenvironment, further supporting blood vessel growth around tumors. The intricate network of communication between neural constituents and the vascular system accentuates the potential of therapeutically targeting neural-mediated pathways as an innovative strategy to modulate tumor angiogenesis and, consequently, neoplastic proliferation. Hereby, we review studies that evaluate the precise molecular interplay and the potential clinical ramifications of manipulating neural elements for the purpose of anti-angiogenic therapeutics within the scope of cancer treatment.
Collapse
Affiliation(s)
- Sharif Shalabi
- Maisonneuve-Rosemont Hospital Research Center, Boulevard de l’Assomption, Montréal, QC, Canada
| | - Ali Belayachi
- Maisonneuve-Rosemont Hospital Research Center, Boulevard de l’Assomption, Montréal, QC, Canada
| | - Bruno Larrivée
- Maisonneuve-Rosemont Hospital Research Center, Boulevard de l’Assomption, Montréal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Montréal, QC, Canada
- Ophthalmology, Université de Montréal, boul. Édouard-Montpetit, Montréal, QC, Canada
| |
Collapse
|
4
|
Paşcalău R, Badea TC. Signaling - transcription interactions in mouse retinal ganglion cells early axon pathfinding -a literature review. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1180142. [PMID: 38983012 PMCID: PMC11182120 DOI: 10.3389/fopht.2023.1180142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/21/2023] [Indexed: 07/11/2024]
Abstract
Sending an axon out of the eye and into the target brain nuclei is the defining feature of retinal ganglion cells (RGCs). The literature on RGC axon pathfinding is vast, but it focuses mostly on decision making events such as midline crossing at the optic chiasm or retinotopic mapping at the target nuclei. In comparison, the exit of RGC axons out of the eye is much less explored. The first checkpoint on the RGC axons' path is the optic cup - optic stalk junction (OC-OS). OC-OS development and the exit of the RGC pioneer axons out of the eye are coordinated spatially and temporally. By the time the optic nerve head domain is specified, the optic fissure margins are in contact and the fusion process is ongoing, the first RGCs are born in its proximity and send pioneer axons in the optic stalk. RGC differentiation continues in centrifugal waves. Later born RGC axons fasciculate with the more mature axons. Growth cones at the end of the axons respond to guidance cues to adopt a centripetal direction, maintain nerve fiber layer restriction and to leave the optic cup. Although there is extensive information on OC-OS development, we still have important unanswered questions regarding its contribution to the exit of the RGC axons out of the eye. We are still to distinguish the morphogens of the OC-OS from the axon guidance molecules which are expressed in the same place at the same time. The early RGC transcription programs responsible for axon emergence and pathfinding are also unknown. This review summarizes the molecular mechanisms for early RGC axon guidance by contextualizing mouse knock-out studies on OC-OS development with the recent transcriptomic studies on developing RGCs in an attempt to contribute to the understanding of human optic nerve developmental anomalies. The published data summarized here suggests that the developing optic nerve head provides a physical channel (the closing optic fissure) as well as molecular guidance cues for the pioneer RGC axons to exit the eye.
Collapse
Affiliation(s)
- Raluca Paşcalău
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- Ophthalmology Clinic, Cluj County Emergency Hospital, Cluj-Napoca, Romania
| | - Tudor Constantin Badea
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- National Center for Brain Research, Institutul de Cercetări pentru Inteligență Artificială, Romanian Academy, Bucharest, Romania
| |
Collapse
|
5
|
Zhou R, Zhou D, Yang S, Shi Z, Pan H, Jin Q, Ding Z. Neurotoxicity of polystyrene nanoplastics with different particle sizes at environment-related concentrations on early zebrafish embryos. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162096. [PMID: 36791853 DOI: 10.1016/j.scitotenv.2023.162096] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/30/2023] [Accepted: 02/04/2023] [Indexed: 06/18/2023]
Abstract
Nanoplastics (NPs) have received global attention due to their wide application and detection in various environmental or biological media. NPs can penetrate physical barriers and accumulate in organisms after being ingested, producing a variety of toxic effects and possessing particle size-dependent effects, distinguishing them from traditional contaminants. This paper explored the neurotoxicity of polystyrene (PS)-NPs of different particle sizes on zebrafish (Danio rerio) embryos at environmental concentrations at the tissue and molecular levels using visualized transgenic zebrafish. Results showed that all particle sizes of PS-NPs produced developmental toxicity in zebrafish embryos and induced neuronal loss, axonal deletion/shortening/hybridization, and developmental and apoptotic-related genetic alterations, ultimately leading to behavioral abnormalities. PS-NPs with smaller sizes may have more severe neurotoxicity due to their entry into the embryo and brain through the chorionic pore before hatching. In addition, PS-NPs at 100 nm and 1000 nm can specifically interfere with GABAergic, cholinergic or serotonergic system and affect neuronal signaling. Our results reveal the neurotoxic risk of NPs, and smaller particle-size NPs may have a greater ecological risk. We anticipate that our study can provide a basis for exploring the toxicity mechanisms of NPs and the environmental risk assessment of NPs.
Collapse
Affiliation(s)
- Ranran Zhou
- School of Environmental Science & Engineering, Nanjing Tech University, 30 Puzhu Southern Road, Nanjing 211816, China
| | - Dao Zhou
- School of Environmental Science & Engineering, Nanjing Tech University, 30 Puzhu Southern Road, Nanjing 211816, China
| | - Shixin Yang
- School of Environmental Science & Engineering, Nanjing Tech University, 30 Puzhu Southern Road, Nanjing 211816, China
| | - Zhiqiao Shi
- School of Environmental Science & Engineering, Nanjing Tech University, 30 Puzhu Southern Road, Nanjing 211816, China
| | - Hui Pan
- School of Environmental Science & Engineering, Nanjing Tech University, 30 Puzhu Southern Road, Nanjing 211816, China
| | - Qijie Jin
- School of Environmental Science & Engineering, Nanjing Tech University, 30 Puzhu Southern Road, Nanjing 211816, China
| | - Zhuhong Ding
- School of Environmental Science & Engineering, Nanjing Tech University, 30 Puzhu Southern Road, Nanjing 211816, China.
| |
Collapse
|
6
|
Sun X, Yang Q, Jing M, Jia X, Tian L, Tao J. Environmentally relevant concentrations of organic (benzophenone-3) and inorganic (titanium dioxide nanoparticles) UV filters co-exposure induced neurodevelopmental toxicity in zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114343. [PMID: 36508829 DOI: 10.1016/j.ecoenv.2022.114343] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 11/20/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
UV filters, widely used in personal care products, are ubiquitous environmental pollutants detected and pose a significant public health concern. Benzophenone-3 (BP3) and titanium dioxide nanoparticles (nano-TiO2) are the predominant organic and inorganic UV filters in environmental media. However, few studies have explored the combined developmental neurotoxic (DNT) effects and the underlying mechanisms when co-exposed to BP3 and nano-TiO2. In the present study, zebrafish (Danio rerio) embryos were exposed to environmentally relevant concentrations of BP3 (10 μg/L), nano-TiO2 (100 μg/L), and mixtures starting from 6 h post fertilization (hpf), respectively. Developmental indicators and motor behaviors were investigated at various developmental stages. Our results showed that BP3 alone or co-exposed with nano-TiO2 increased spontaneous movement at 24 hpf, co-exposure decreased touch response at 30 hpf and hatching rate at 60 hpf. Consistent with these motor deficits, co-exposure to BP3 and nano-TiO2 inhibited relative axon length of primary motor neuron during the early developmental stages, disturbed the expression of axonal growth-related genes at 30 and 48 hpf, increased cell apoptosis on the head region and mRNA levels of apoptosis-related genes, and also increased reactive oxygen species (ROS) levels in zebrafish, suggesting the functional relevance of structural changes. Taken together, our findings demonstrated that BP3 alone or in combination with nano-TiO2 at environmentally relevant concentrations induced evident neurotoxic effects on the developing embryos in zebrafish.
Collapse
Affiliation(s)
- Xiaowei Sun
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China
| | - Qinyuan Yang
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China
| | - Min Jing
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China
| | - Xinrui Jia
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China
| | - Linxuan Tian
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China
| | - Junyan Tao
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|
7
|
Ros O, Nicol X. Axon pathfinding and targeting: (R)evolution of insights from in vitro assays. Neuroscience 2023; 508:110-122. [PMID: 36096337 DOI: 10.1016/j.neuroscience.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 01/17/2023]
Abstract
Investigating axonal behaviors while neurons are connecting with each other has been a challenge since the early studies on nervous system development. While molecule-driven axon pathfinding has been theorized by observing neurons at different developmental stages in vivo, direct observation and measurements of axon guidance behaviors required the invention of in vitro systems enabling to test the impact of molecules or cellular extracts on axons growing in vitro. With time, the development of novel in vivo approaches has confirmed the mechanisms highlighted in culture and has led in vitro systems to be adapted for cellular processes that are still inaccessible in intact organisms. We here review the evolution of these in vitro assays, which started with crucial contributions from the Bonhoeffer lab.
Collapse
Affiliation(s)
- Oriol Ros
- Universitat de Barcelona, Department of Cell Biology, Physiology and Immunology, Avinguda Diagonal 643, 08028 Barcelona, Catalonia, Spain
| | - Xavier Nicol
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
8
|
Yu H, Zhang J, Chen Y, Chen J, Qiu Y, Zhao Y, Li H, Xia S, Chen S, Zhu J. The adverse effects of fluxapyroxad on the neurodevelopment of zebrafish embryos. CHEMOSPHERE 2022; 307:135751. [PMID: 35863420 DOI: 10.1016/j.chemosphere.2022.135751] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023]
Abstract
Fluxapyroxad (Flu), one of the succinate dehydrogenase-inhibited (SDHI) fungicides, has been extensively used in crop fungal disease control. Despite its increasing use in modern agriculture and long-term retention in the environment, the potentially toxic effects of Flu in vivo, especially on neurodevelopment, remain under-evaluated. In this study, zebrafish embryos were exposed to Flu at concentrations of 0.5, 0.75, and 1 mg/L for 96 h to evaluate the neurotoxicity of Flu. The results showed that Flu caused concentration-dependent malformations, including shorter body length, smaller head and eyes, and yolk sac edema. After exposure to Flu, larval zebrafish exhibited severe motor aberrations. Flu at a concentration of 1 mg/L significantly decreased dopamine level and notably altered acetylcholinesterase (AChE) activity and acetylcholine (ACh) content. Abnormal central nervous system (CNS) neurogenesis and disordered motor neuron development were observed in Tg (HUC-GFP) and Tg (hb9-GFP) zebrafish in Flu-treated groups. The expression of key genes involved in neurotransmission and neurodevelopment further proved that Flu impaired the zebrafish nervous system. This work contributes to our understanding of the neurotoxic effects and mechanisms induced by Flu in zebrafish and may help us take precautions against the neurotoxicity of Flu.
Collapse
Affiliation(s)
- Huilin Yu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Junhui Zhang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yinghong Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Juan Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yang Qiu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yan Zhao
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Honghao Li
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Shengyao Xia
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Jiajin Zhu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
9
|
Young TL, Whisenhunt KN, LaMartina SM, Hewitt AW, Mackey DA, Tompson SW. Sonic Hedgehog Intron Variant Associated With an Unusual Pediatric Cortical Cataract. Invest Ophthalmol Vis Sci 2022; 63:25. [PMID: 35749127 PMCID: PMC9234370 DOI: 10.1167/iovs.63.6.25] [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] [Indexed: 11/24/2022] Open
Abstract
Purpose To identify the genetic basis of an unusual pediatric cortical cataract demonstrating autosomal dominant inheritance in a large European–Australian pedigree. Methods DNA from four affected individuals were exome sequenced utilizing a NimbleGen SeqCap EZ Exome V3 kit and HiSeq 2500. DNA from 12 affected and four unaffected individuals were genotyped using Human OmniExpress-24 BeadChips. Multipoint linkage and haplotyping were performed (Superlink-Online SNP). DNA from one affected individual and his unaffected father were whole-genome sequenced on a HiSeq X Ten system. Rare small insertions/deletions and single-nucleotide variants (SNVs) were identified in the disease-linked region (Golden Helix SVS). Combined Annotation Dependent Depletion (CADD) analysis predicted variant deleteriousness. Putative enhancer function and variant effects were determined using the Dual-Glo Luciferase Assay system. Results Linkage mapping identified a 6.23-centimorgan support interval at chromosome 7q36. A co-segregating haplotype refined the critical region to 6.03 Mbp containing 21 protein-coding genes. Whole-genome sequencing uncovered 114 noncoding variants from which CADD predicted one was highly deleterious, a novel substitution within intron-1 of the sonic hedgehog signaling molecule (SHH) gene. ENCODE data suggested this site was a putative enhancer, subsequently confirmed by luciferase reporter assays with variant-associated gene overexpression. Conclusions In a large pedigree, we have identified a SHH intron variant that co-segregates with an unusual pediatric cortical cataract phenotype. SHH is important for lens formation, and mutations in its receptor (PTCH1) cause syndromic cataract. Our data implicate increased function of an enhancer important for SHH expression primarily within developing eye tissues.
Collapse
Affiliation(s)
- Terri L Young
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Kristina N Whisenhunt
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Sarah M LaMartina
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Alex W Hewitt
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia.,Lions Eye Institute, Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia.,Eye Department, Royal Hobart Hospital, University of Tasmania, Hobart, Tasmania, Australia
| | - David A Mackey
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia.,Lions Eye Institute, Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia.,Eye Department, Royal Hobart Hospital, University of Tasmania, Hobart, Tasmania, Australia
| | - Stuart W Tompson
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| |
Collapse
|
10
|
Ahmed G, Shinmyo Y. Multiple Functions of Draxin/Netrin-1 Signaling in the Development of Neural Circuits in the Spinal Cord and the Brain. Front Neuroanat 2021; 15:766911. [PMID: 34899198 PMCID: PMC8655782 DOI: 10.3389/fnana.2021.766911] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/22/2021] [Indexed: 11/30/2022] Open
Abstract
Axon guidance proteins play key roles in the formation of neural circuits during development. We previously identified an axon guidance cue, named draxin, that has no homology with other axon guidance proteins. Draxin is essential for the development of various neural circuits including the spinal cord commissure, corpus callosum, and thalamocortical projections. Draxin has been shown to not only control axon guidance through netrin-1 receptors, deleted in colorectal cancer (Dcc), and neogenin (Neo1) but also modulate netrin-1-mediated axon guidance and fasciculation. In this review, we summarize the multifaceted functions of draxin and netrin-1 signaling in neural circuit formation in the central nervous system. Furthermore, because recent studies suggest that the distributions and functions of axon guidance cues are highly regulated by glycoproteins such as Dystroglycan and Heparan sulfate proteoglycans, we discuss a possible function of glycoproteins in draxin/netrin-1-mediated axon guidance.
Collapse
Affiliation(s)
- Giasuddin Ahmed
- Department of Neuroscience and Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
11
|
Moreau N, Boucher Y. Hedging against Neuropathic Pain: Role of Hedgehog Signaling in Pathological Nerve Healing. Int J Mol Sci 2020; 21:ijms21239115. [PMID: 33266112 PMCID: PMC7731127 DOI: 10.3390/ijms21239115] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 11/28/2020] [Accepted: 11/29/2020] [Indexed: 12/23/2022] Open
Abstract
The peripheral nervous system has important regenerative capacities that regulate and restore peripheral nerve homeostasis. Following peripheral nerve injury, the nerve undergoes a highly regulated degeneration and regeneration process called Wallerian degeneration, where numerous cell populations interact to allow proper nerve healing. Recent studies have evidenced the prominent role of morphogenetic Hedgehog signaling pathway and its main effectors, Sonic Hedgehog (SHH) and Desert Hedgehog (DHH) in the regenerative drive following nerve injury. Furthermore, dysfunctional regeneration and/or dysfunctional Hedgehog signaling participate in the development of chronic neuropathic pain that sometimes accompanies nerve healing in the clinical context. Understanding the implications of this key signaling pathway could provide exciting new perspectives for future research on peripheral nerve healing.
Collapse
Affiliation(s)
- Nathan Moreau
- Department of Oral Medicine and Oral Surgery, Bretonneau Hospital (AP-HP), 75018 Paris, France;
- Faculty of Dental Medicine-Montrouge, University of Paris, 92120 Montrouge, France
| | - Yves Boucher
- Department of Dental Medicine, Pitié-Salpêtrière Hospital (AP-HP), 75013 Paris, France
- Faculty of Dental Medicine-Garancière, University of Paris, 75006 Paris, France
- Correspondence:
| |
Collapse
|
12
|
Guo SY, Zhang Y, Zhu XY, Zhou JL, Li J, Li CQ, Wu LR. Developmental neurotoxicity and toxic mechanisms induced by olaquindox in zebrafish. J Appl Toxicol 2020; 41:549-560. [PMID: 33111391 DOI: 10.1002/jat.4062] [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: 04/29/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 12/30/2022]
Abstract
Olaquindox (OLA) has been widely used as an animal feed additive in China for decades; however, its toxicity and toxic mechanisms have not been well investigated. In this study, the developmental neurotoxicity and toxic mechanisms of OLA were evaluated in zebrafish. Zebrafish embryos were exposed to different concentrations of OLA (25-1,000 mg/L) from 6 to 120 hours post fertilization (hpf). OLA exposure resulted in many abnormal phenotypes in zebrafish, including shortened body length, notochord degeneration, spinal curvature, brain apoptosis, damage of axon and peripheral motor neuron, and hepatotoxicity. Interestingly, OLA increased zebrafish spontaneous tail coiling, while reduced locomotor capacity. Quantitative polymerase chain reaction (Q-PCR) showed that the expression levels of nine marker genes for nervous system functions or development, namely, α1-tubulin, glial fibrillary acidic protein (gfap), myelin basic protein (mbp), synapsinII a (syn2a), sonic hedgehog a (shha), encoding HuC (elavl3), mesencephalic astrocyte-derived neurotrophic factor (manf) growth associated protein 43 (gap43), and acetylcholinesterase (ache) were all down-regulated significantly in zebrafish after treated with OLA. Besides, the anti-apoptotic and pro-apoptotic genes bcl-2/bax ratio was reduced. These results show that OLA exposure could cause severe developmental neurotoxicity in the early stages of zebrafish life and OLA might induce neurotoxicity by inhibiting the expression of neuro-developmental genes and promoting apoptosis.
Collapse
Affiliation(s)
- Sheng-Ya Guo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Binwen Road, Hangzhou, 310053, China.,Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China
| | - Yong Zhang
- Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China
| | - Xiao-Yu Zhu
- Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China
| | - Jia-Li Zhou
- Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China
| | - Jiao Li
- Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China
| | - Chun-Qi Li
- Research and Development Department, Hunter Biotechnology, Inc., Jiangling Road, Hangzhou, 310051, China.,Research and Development Department, New Hunter Testing and Technology Co., Ltd, Xinjinhu Road, Nanjing, 210046, China
| | - Li-Ren Wu
- Laboratory Animal Regulatory Center, Hangzhou Medical College, Tianmushan Road, Hangzhou, 310013, China
| |
Collapse
|
13
|
Kim Y, Lee J, Seppala M, Cobourne MT, Kim SH. Ptch2/Gas1 and Ptch1/Boc differentially regulate Hedgehog signalling in murine primordial germ cell migration. Nat Commun 2020; 11:1994. [PMID: 32332736 PMCID: PMC7181751 DOI: 10.1038/s41467-020-15897-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 04/01/2020] [Indexed: 12/24/2022] Open
Abstract
Gas1 and Boc/Cdon act as co-receptors in the vertebrate Hedgehog signalling pathway, but the nature of their interaction with the primary Ptch1/2 receptors remains unclear. Here we demonstrate, using primordial germ cell migration in mouse as a developmental model, that specific hetero-complexes of Ptch2/Gas1 and Ptch1/Boc mediate the process of Smo de-repression with different kinetics, through distinct modes of Hedgehog ligand reception. Moreover, Ptch2-mediated Hedgehog signalling induces the phosphorylation of Creb and Src proteins in parallel to Gli induction, identifying a previously unknown Ptch2-specific signal pathway. We propose that although Ptch1 and Ptch2 functionally overlap in the sequestration of Smo, the spatiotemporal expression of Boc and Gas1 may determine the outcome of Hedgehog signalling through compartmentalisation and modulation of Smo-downstream signalling. Our study identifies the existence of a divergent Hedgehog signal pathway mediated by Ptch2 and provides a mechanism for differential interpretation of Hedgehog signalling in the germ cell niche. How co-receptors Gas1 and Boc interact with Ptch1/2 receptors and regulate Hh signalling is unclear. Here, the authors demonstrate that the spatiotemporal expression of Gas1 and Boc determines how Hh signalling affects the dynamic migration of murine primordial germ cells.
Collapse
Affiliation(s)
- Yeonjoo Kim
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Jiyoung Lee
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Maisa Seppala
- Centre for Craniofacial and Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences King's College London Floor 27, Guy's Hospital, London, SE1 9RT, UK
| | - Martyn T Cobourne
- Centre for Craniofacial and Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences King's College London Floor 27, Guy's Hospital, London, SE1 9RT, UK
| | - Soo-Hyun Kim
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, UK.
| |
Collapse
|
14
|
Sonic Hedgehog upregulation does not enhance the survival and engraftment of stem cell-derived cardiomyocytes in infarcted hearts. PLoS One 2020; 15:e0227780. [PMID: 31945113 PMCID: PMC6964843 DOI: 10.1371/journal.pone.0227780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/28/2019] [Indexed: 01/02/2023] Open
Abstract
The engraftment of human stem cell-derived cardiomyocytes (hSC-CMs) is a promising treatment for remuscularizing the heart wall post-infarction, but it is plagued by low survival of transplanted cells. We hypothesize that this low survival rate is due to continued ischemia within the infarct, and that increasing the vascularization of the scar will ameliorate the ischemia and improve hSC-CM survival and engraftment. An adenovirus expressing the vascular growth factor Sonic Hedgehog (Shh) was injected into the infarcted myocardium of rats immediately after ischemia/reperfusion, four days prior to hSC-CM injection. By two weeks post-cell injection, Shh treatment had successfully increased capillary density outside the scar, but not within the scar. In addition, there was no change in vessel size or percent vascular volume when compared to cell injection alone. Micro-computed tomography revealed that Shh failed to increase the number and size of larger vessels. It also had no effect on graft size or heart function when compared to cell engraftment alone. Our data suggests that, when combined with the engraftment of hSC-CMs, expression of Shh within the infarct scar and surrounding myocardium is unable to increase vascularization of the infarct scar, and it does not improve survival or function of hSC-CM grafts.
Collapse
|
15
|
Herrera E, Agudo-Barriuso M, Murcia-Belmonte V. Cranial Pair II: The Optic Nerves. Anat Rec (Hoboken) 2018; 302:428-445. [DOI: 10.1002/ar.23922] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/19/2017] [Accepted: 05/14/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Eloísa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina; Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca); Murcia Spain
| | - Verónica Murcia-Belmonte
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| |
Collapse
|
16
|
Liu M, Chen X, Liu H, Di Y. Expression and significance of the Hedgehog signal transduction pathway in oxygen-induced retinal neovascularization in mice. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:1337-1346. [PMID: 29861625 PMCID: PMC5968796 DOI: 10.2147/dddt.s149594] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aim The aim of the study was to investigate the signal transduction mechanism of Hedgehog–vascular endothelial growth factor in oxygen-induced retinopathy (OIR) and the effects of cyclopamine on OIR. Methods An OIR model was established in C57BL/6J mice exposed to hyperoxia. Two hundred mice were randomly divided into a control group, an OIR group, an OIR-control group (treated with isometric phosphate-buffered saline by intravitreal injection), and a cyclopamine group (treated with cyclopamine by intravitreal injection), with 50 mice in each group. The retinal vascular morphology was observed using adenosine diphosphatase and number counting using hematoxylin and eosin-stained image. Quantitative real-time quantitative polymerase chain reaction was used to detect mRNA expression. Protein location and expression were evaluated using immunohistochemistry and Western blot. Results The OIR group and OIR-control group demonstrated large-area pathological neovascularization and nonperfused area when compared with the control group (both P<0.05). The area of nonperfusion and neovascularization in the cyclopamine group was significantly reduced compared with the OIR and OIR-control groups (both P<0.05). Compared with the control group, the OIR and OIR-control groups had more vascular endothelial cells breaking through the inner limiting membrane. The number of new blood vessel endothelial cell nuclei in the cyclopamine group was significantly reduced (both P<0.05) when compared with the OIR and OIR-control groups. The mRNA and protein expressions of Smoothened, Gli1, and vascular endothelial growth factor in the signal pathway of the OIR and OIR-control groups were significantly higher than those of the control group; however, in the cyclopamine group, these factors were reduced when compared with the OIR and OIR-control groups (all P<0.05). Conclusion Our data suggest that abnormal expression of the Hedgehog signaling pathway may be closely associated with the formation of OIR. Inhibiting the Smoothened receptor using cyclopamine could control retinal neovascularization, providing new ideas and measures for the prevention of oxygen-induced retinal neovascularization.
Collapse
Affiliation(s)
- Meilin Liu
- Department of Ophthalmology, Shengjing Affiliated Hospital, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaolong Chen
- Department of Ophthalmology, Shengjing Affiliated Hospital, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Henan Liu
- Department of Ophthalmology, Shengjing Affiliated Hospital, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yu Di
- Department of Ophthalmology, Shengjing Affiliated Hospital, China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
17
|
Tan CL, Sheard PW, Jasoni CL. Developing neurites from mouse basal forebrain gonadotropin-releasing hormone neurons use Sonic hedgehog to modulate their growth. Int J Dev Neurosci 2018; 68:89-97. [PMID: 29787797 DOI: 10.1016/j.ijdevneu.2018.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/14/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Hypothalamic gonadotropin-releasing hormone (GnRH) neurons are required for fertility in all mammalian species studied to date. GnRH neuron cell bodies reside in the basal forebrain, and most extend long neurites in the caudal direction to terminate at the median eminence (ME), the site of hormone secretion. Using in vitro neurite growth assays, histological methods, and genetic deletion strategies in mice we have analysed the role of the morphogen and neurite growth and guidance molecule, Sonic hedgehog (Shh), in the growth of GnRH neurites to their target. Immunohistochemistry revealed that Shh was present in the basal forebrain, the preoptic area (POA) and mediobasal hypothalamus (MBH) at gestational day 14.5 (GD 14.5), a time when GnRH neurites grow towards the ME. Furthermore, in situ hybridization revealed that mRNA encoding the Shh receptor, Smoothened (Smo), was present in GnRH neurons from GD 15.5, when the first GnRH neurites are extending towards the MBH. In vitro neurite growth assays using hypothalamic explants from GD 15.5 fetuses in 3-D collagen gels showed that Shh was able to significantly stimulate GnRH neurite outgrowth. Finally, genetic deletion of Smo specifically from GnRH neurons in vivo, using Cre-loxP technology, resulted in a significant decrease in GnRH neurites innervating the ME. These experiments demonstrate that GnRH neurites use Shh for their neurite development, provide further understanding of the mechanisms by which GnRH nerve terminals arrive at their site of hormone secretion, and identify an additional hypothalamic neuronal population for which Shh/Smo signaling is developmentally important.
Collapse
Affiliation(s)
- C L Tan
- Department of Anatomy, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand; Centre for Neuroendocrinology, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand.
| | - P W Sheard
- Department of Physiology, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand.
| | - C L Jasoni
- Department of Anatomy, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand; Centre for Neuroendocrinology, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand.
| |
Collapse
|
18
|
Jiang F, Liu J, Zeng X, Yu L, Liu C, Wang J. Tris (2-butoxyethyl) phosphate affects motor behavior and axonal growth in zebrafish (Danio rerio) larvae. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 198:215-223. [PMID: 29558706 DOI: 10.1016/j.aquatox.2018.03.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 06/08/2023]
Abstract
Tris (2-butoxyethyl) phosphate (TBOEP) is an environmental contaminant that poses serious risks to aquatic organisms and their associated ecosystem. Recently, the reproductive and developmental toxicology of TBOEP has been reported. However, fewer studies have assessed the neurotoxic effects in zebrafish (Danio rerio) larvae. In this study, zebrafish embryos were subjected to waterborne exposure of TBOEP at 0, 50, 500, 1500 and 2500 μg/L from 2 to 144-h post-fertilization (hpf). Behavioral measurements showed that TBOEP exposure reduced embryonic spontaneous movement and decreased swimming speed of larvae in response to dark stimulation. In accordance with these motor effects, TBOEP treatment reduced neuron-specific GFP expression in transgenic Tg (HuC-GFP) zebrafish larvae and inhibited the growth of secondary motoneurons, as well as decreased expression of marker genes related to central nervous system development in TBOEP treated group. Furthermore, increased concentrations of reactive oxygen species (ROS) and malondialdehyde (MDA), as well as reduction of SOD activity were detected in TBOEP exposure group. The present results showed that the alteration in motor neuron and oxidative stress could together lead to the motor behavior alterations induced by TBOEP.
Collapse
Affiliation(s)
- Fan Jiang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Jue Liu
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Xinyue Zeng
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Liqin Yu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Chunsheng Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianghua Wang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
19
|
Zhu B, Zhao G, Yang L, Zhou B. Tetrabromobisphenol A caused neurodevelopmental toxicity via disrupting thyroid hormones in zebrafish larvae. CHEMOSPHERE 2018; 197:353-361. [PMID: 29407805 DOI: 10.1016/j.chemosphere.2018.01.080] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/14/2018] [Accepted: 01/16/2018] [Indexed: 05/27/2023]
Abstract
Tetrabromobisphenol A (TBBPA), one of the most widely used brominated flame retardants (BFRs), has resulted in worldwide environmental contamination. TBBPA has been reported as a thyroid endocrine disruptor and a potential neurotoxicant. However, the underlying mechanism is still not clear. In this study, zebrafish (Danio rerio) embryos (2 h post-fertilization, hpf) were exposed to different concentrations of TBBPA (50, 100, 200 and 400 μg/L) alone or in combination with 3,3',5-triiodo-l-thyronine (T3, 20 μg/L + TBBPA, 200 μg/L). The results confirmed that TBBPA could evoke thyroid disruption by observations of increased T4 contents and decreased T3 contents, accompanied by up-regulated tshβ, tg mRNA and down-regulated ttr and trβ mRNA levels in zebafish larvae. TBBPA-induced neurodevelopmental toxicity was also indicated by down-regulated transcription of genes related to central nervous system (CNS) development (e.g., α1-tubulin, mbp and shha), and decreased locomotor activity and average swimming speed. Our results further demonstrated that treatment with T3 could reverse or eliminate TBBPA-induced effects on thyroidal and neurodevelopmental parameters. Given the above, we hypothesize that the observed neurodevelopmental toxicity in the present study could be attributed to the thyroid hormone disruptions by TBBPA.
Collapse
Affiliation(s)
- Biran Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; Department of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Gang Zhao
- Department of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Lihua Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| |
Collapse
|
20
|
Peng J, Fabre PJ, Dolique T, Swikert SM, Kermasson L, Shimogori T, Charron F. Sonic Hedgehog Is a Remotely Produced Cue that Controls Axon Guidance Trans-axonally at a Midline Choice Point. Neuron 2018; 97:326-340.e4. [DOI: 10.1016/j.neuron.2017.12.028] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 11/10/2017] [Accepted: 12/15/2017] [Indexed: 12/11/2022]
|
21
|
Cheng R, Jia Y, Dai L, Liu C, Wang J, Li G, Yu L. Tris(1,3-dichloro-2-propyl) phosphate disrupts axonal growth, cholinergic system and motor behavior in early life zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 192:7-15. [PMID: 28898785 DOI: 10.1016/j.aquatox.2017.09.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/30/2017] [Accepted: 09/02/2017] [Indexed: 06/07/2023]
Abstract
Tris(1,3-dichloro-2-propyl) phosphate (TDCIPP) could have neurotoxic effects and alter motor behaviors in zebrafish (Danio rerio) larvae, however, the underlying mechanisms are still unknown. In this study, zebrafish embryos were subjected to waterborne exposure of TDCIPP at 100, 300, 600, 900μg/L from 2 to 120-h post-fertilization (hpf). Behavioral measurements indicate that TDCIPP exposure significantly elevated spontaneous movement, and altered swimming behavior response of larvae to both light and dark stimulation. Interestingly, in accordance with these motor effects, TDCIPP significantly decreased expression of the neuron-specific GFP in transgenic (HuC-GFP) zebrafish larvae as well as decreased expression of the neural marker genes elavl3 and ngn1, inhibited the axonal growth of the secondary motoneurons and altered the expressions of axon-related genes (α1-tubulin, shha and netrin2) in zebrafish larvae. Furthermore, TDCIPP exposure at 900μg/L significantly increased the activity of acetylcholinesterase (AChE) enzyme, and decreased the total acetylcholine (ACh) concentration. Our data indicate that the alteration in motor neuron and inhibition of cholinergic system could together lead to the TDCIPP induced motor behavior alterations in zebrafish larvae.
Collapse
Affiliation(s)
- Rui Cheng
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Yali Jia
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Lili Dai
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, China
| | - Chunsheng Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Hunan Changde 415000, China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, China
| | - Jianghua Wang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Guangyu Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Liqin Yu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
22
|
Bhattacharjee N, Folch A. Large-scale microfluidic gradient arrays reveal axon guidance behaviors in hippocampal neurons. MICROSYSTEMS & NANOENGINEERING 2017; 3:17003. [PMID: 31057858 PMCID: PMC6445017 DOI: 10.1038/micronano.2017.3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/30/2016] [Accepted: 12/10/2016] [Indexed: 05/05/2023]
Abstract
High-throughput quantitative approaches to study axon growth behaviors have remained a challenge. We have developed a 1024-chamber microfluidic gradient generator array that enables large-scale investigations of axon guidance and growth dynamics from individual primary mammalian neurons, which are exposed to gradients of diffusible molecules. Our microfluidic method (a) generates statistically rich data sets, (b) produces a stable, reproducible gradient with negligible shear stresses on the culture surface, (c) is amenable to the long-term culture of primary neurons without any unconventional protocol, and (d) eliminates the confounding influence of cell-secreted factors. Using this platform, we demonstrate that hippocampal axon guidance in response to a netrin-1 gradient is concentration-dependent-attractive at higher concentrations and repulsive at lower concentrations. We also show that the turning of the growth cone depends on the angle of incidence of the gradient. Our study highlights the potential of microfluidic devices in producing large amounts of data from morphogen and chemokine gradients that play essential roles not only in axonal navigation but also in stem cell differentiation, cell migration, and immune response.
Collapse
Affiliation(s)
- Nirveek Bhattacharjee
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Foege N423-A, Seattle, WA 98195, USA
- ()
| | - Albert Folch
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Foege N423-A, Seattle, WA 98195, USA
| |
Collapse
|
23
|
The Many Hats of Sonic Hedgehog Signaling in Nervous System Development and Disease. J Dev Biol 2016; 4:jdb4040035. [PMID: 29615598 PMCID: PMC5831807 DOI: 10.3390/jdb4040035] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/17/2016] [Accepted: 11/29/2016] [Indexed: 02/06/2023] Open
Abstract
Sonic hedgehog (Shh) signaling occurs concurrently with the many processes that constitute nervous system development. Although Shh is mostly known for its proliferative and morphogenic action through its effects on neural stem cells and progenitors, it also contributes to neuronal differentiation, axonal pathfinding and synapse formation and function. To participate in these diverse events, Shh signaling manifests differently depending on the maturational state of the responsive cell, on the other signaling pathways regulating neural cell function and the environmental cues that surround target cells. Shh signaling is particularly dynamic in the nervous system, ranging from canonical transcription-dependent, to non-canonical and localized to axonal growth cones. Here, we review the variety of Shh functions in the developing nervous system and their consequences for neurodevelopmental diseases and neural regeneration, with particular emphasis on the signaling mechanisms underlying Shh action.
Collapse
|
24
|
Kahn BM, Corman TS, Lovelace K, Hong M, Krauss RS, Epstein DJ. Prenatal ethanol exposure in mice phenocopies Cdon mutation by impeding Shh function in the etiology of optic nerve hypoplasia. Dis Model Mech 2016; 10:29-37. [PMID: 27935818 PMCID: PMC5278523 DOI: 10.1242/dmm.026195] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 11/16/2016] [Indexed: 01/01/2023] Open
Abstract
Septo-optic dysplasia (SOD) is a congenital disorder characterized by optic nerve, pituitary and midline brain malformations. The clinical presentation of SOD is highly variable with a poorly understood etiology. The majority of SOD cases are sporadic, but in rare instances inherited mutations have been identified in a small number of transcription factors, some of which regulate the expression of Sonic hedgehog (Shh) during mouse forebrain development. SOD is also associated with young maternal age, suggesting that environmental factors, including alcohol consumption at early stages of pregnancy, might increase the risk of developing this condition. Here, we address the hypothesis that SOD is a multifactorial disorder stemming from interactions between mutations in Shh pathway genes and prenatal ethanol exposure. Mouse embryos with mutations in the Shh co-receptor, Cdon, were treated in utero with ethanol or saline at embryonic day 8 (E8.0) and evaluated for optic nerve hypoplasia (ONH), a prominent feature of SOD. We show that both Cdon-/- mutation and prenatal ethanol exposure independently cause ONH through a similar pathogenic mechanism that involves selective inhibition of Shh signaling in retinal progenitor cells, resulting in their premature cell-cycle arrest, precocious differentiation and failure to properly extend axons to the optic nerve. The ONH phenotype was not exacerbated in Cdon-/- embryos treated with ethanol, suggesting that an intact Shh signaling pathway is required for ethanol to exert its teratogenic effects. These results support a model whereby mutations in Cdon and prenatal ethanol exposure increase SOD risk through spatiotemporal perturbations in Shh signaling activity.
Collapse
Affiliation(s)
- Benjamin M Kahn
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Tanya S Corman
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Korah Lovelace
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Mingi Hong
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Robert S Krauss
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Douglas J Epstein
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
25
|
On the mechanism of smell loss in patients with Type II congenital hyposmia. Am J Otolaryngol 2016; 37:436-41. [PMID: 27221029 DOI: 10.1016/j.amjoto.2016.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 11/23/2022]
Abstract
BACKGROUND Smell function has been initiated with theophylline treatment in 63% of patients with Type II congenital smell loss. Based upon a systematic evaluation of the protein components of nasal mucus we have demonstrated that interactions among four chemical moieties in nasal mucus may play significant roles in this initiation. Prior to treatment three of these moieties, cAMP, cGMP and sonic hedgehog (Shh), were significantly decreased in concentration whereas one of these moieties, TNFalpha, was increased in concentration. The mechanism(s) responsible for initiation of smell function in these patients, not immediately apparent, may depend upon understanding interactions among these moieties. METHODS Measurements of cAMP, cGMP, Shh and TNFalpha in nasal mucus by specific spectrophotometric immunoassays before and after treatment with theophylline. RESULTS Before theophylline treatment cAMP, cGMP and Shh in nasal mucus, which act as growth factors to support olfactory receptor function, were significantly decreased below normal levels whereas TNFalpha which acts as a "death factor" to inhibit olfactory receptor function was significantly increased above normal. After theophylline treatment cAMP, cGMP and Shh increased significantly whereas TNFalpha decreased significantly. CONCLUSIONS These results indicate that there are specific biochemical changes associated with smell loss in patients with Type II congenital smell loss and that correction of these biochemical changes are associated with initiation of smell function in these patients. Understanding these relationships play an important role in understanding receptor action in smell function.
Collapse
|
26
|
Wu Q, Yan W, Liu C, Li L, Yu L, Zhao S, Li G. Microcystin-LR exposure induces developmental neurotoxicity in zebrafish embryo. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2016; 213:793-800. [PMID: 27038211 DOI: 10.1016/j.envpol.2016.03.048] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/18/2016] [Accepted: 03/18/2016] [Indexed: 06/05/2023]
Abstract
Microcystin-LR (MCLR) is a commonly acting potent hepatotoxin and has been pointed out of potentially causing developmental neurotoxicity, but the exact mechanism is little known. In this study, zebrafish embryos were exposed to 0, 0.8, 1.6 or 3.2 mg/L MCLR for 120 h. MCLR exposure through submersion caused serious hatching delay and body length decrease. The content of MCLR in zebrafish larvae was analyzed and the results demonstrated that MCLR can accumulate in zebrafish larvae. The locomotor speed of zebrafish larvae was decreased. Furthermore, the dopamine and acetylcholine (ACh) content were detected to be significantly decreased in MCLR exposure groups. And the acetylcholinesterase (AChE) activity was significantly increased after exposure to 1.6 and 3.2 mg/L MCLR. The transcription pattern of manf, chrnα7 and ache gene was consistent with the change of the dopamine content, ACh content and AChE activity. Gene expression involved in the development of neurons was also measured. ɑ1-tubulin and shha gene expression were down-regulated, whereas mbp and gap43 gene expression were observed to be significantly up-regulated upon exposure to MCLR. The above results indicated that MCLR-induced developmental toxicity might attribute to the disorder of cholinergic system, dopaminergic signaling, and the development of neurons.
Collapse
Affiliation(s)
- Qin Wu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan 430070, PR China
| | - Wei Yan
- Institute of Agricultural Quality Standards & Testing Technology, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Chunsheng Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Li Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan 430070, PR China
| | - Liqin Yu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Sujuan Zhao
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Guangyu Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan 430070, PR China.
| |
Collapse
|
27
|
Draxin from neocortical neurons controls the guidance of thalamocortical projections into the neocortex. Nat Commun 2015; 6:10232. [PMID: 26659141 PMCID: PMC4682175 DOI: 10.1038/ncomms10232] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/19/2015] [Indexed: 11/21/2022] Open
Abstract
The thalamocortical tract carries sensory information to the neocortex. It has long been recognized that the neocortical pioneer axons of subplate neurons are essential for thalamocortical development. Herein we report that an axon guidance cue, draxin, is expressed in early-born neocortical neurons, including subplate neurons, and is necessary for thalamocortical development. In draxin−/− mice, thalamocortical axons do not enter the neocortex. This phenotype is sufficiently rescued by the transgenic expression of draxin in neocortical neurons. Genetic interaction data suggest that draxin acts through Deleted in colorectal cancer (DCC) and Neogenin (Neo1), to regulate thalamocortical projections in vivo. Draxin promotes the outgrowth of thalamic axons in vitro and this effect is abolished in thalamic neurons from Dcc and Neo1 double mutants. These results suggest that draxin from neocortical neurons controls thalamocortical projections into the neocortex, and that this effect is mediated through the DCC and Neo1 receptors. During neural development thalamocortical axons follow corticofugal projections into the neocortex. Here, using a combination of knock down and rescue experiments, the authors show that Draxin expression in neocortical cells promotes thalamic axon projections from the internal capsule.
Collapse
|
28
|
Liu Y, Feng J, Li J, Zhao H, Ho TV, Chai Y. An Nfic-hedgehog signaling cascade regulates tooth root development. Development 2015; 142:3374-82. [PMID: 26293299 DOI: 10.1242/dev.127068] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/31/2015] [Indexed: 01/09/2023]
Abstract
Coordination between the Hertwig's epithelial root sheath (HERS) and apical papilla (AP) is crucial for proper tooth root development. The hedgehog (Hh) signaling pathway and Nfic are both involved in tooth root development; however, their relationship has yet to be elucidated. Here, we establish a timecourse of mouse molar root development by histological staining of sections, and we demonstrate that Hh signaling is active before and during root development in the AP and HERS using Gli1 reporter mice. The proper pattern of Hh signaling activity in the AP is crucial for the proliferation of dental mesenchymal cells, because either inhibition with Hh inhibitors or constitutive activation of Hh signaling activity in transgenic mice leads to decreased proliferation in the AP and shorter roots. Moreover, Hh activity is elevated in Nfic(-/-) mice, a root defect model, whereas RNA sequencing and in situ hybridization show that the Hh attenuator Hhip is downregulated. ChIP and RNAscope analyses suggest that Nfic binds to the promoter region of Hhip. Treatment of Nfic(-/-) mice with Hh inhibitor partially restores cell proliferation, AP growth and root development. Taken together, our results demonstrate that an Nfic-Hhip-Hh signaling pathway is crucial for apical papilla growth and proper root formation. This discovery provides insight into the molecular mechanisms regulating tooth root development.
Collapse
Affiliation(s)
- Yang Liu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA Department of Prosthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing 100081, People's Republic of China
| | - Jifan Feng
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Jingyuan Li
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing 100050, People's Republic of China
| | - Hu Zhao
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
29
|
Secreted frizzled related proteins modulate pathfinding and fasciculation of mouse retina ganglion cell axons by direct and indirect mechanisms. J Neurosci 2015; 35:4729-40. [PMID: 25788689 DOI: 10.1523/jneurosci.3304-13.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Retina ganglion cell (RGC) axons grow along a stereotyped pathway undergoing coordinated rounds of fasciculation and defasciculation, which are critical to establishing proper eye-brain connections. How this coordination is achieved is poorly understood, but shedding of guidance cues by metalloproteinases is emerging as a relevant mechanism. Secreted Frizzled Related Proteins (Sfrps) are multifunctional proteins, which, among others, reorient RGC growth cones by regulating intracellular second messengers, and interact with Tolloid and ADAM metalloproteinases, thereby repressing their activity. Here, we show that the combination of these two functions well explain the axon guidance phenotype observed in Sfrp1 and Sfrp2 single and compound mouse mutant embryos, in which RGC axons make subtle but significant mistakes during their intraretinal growth and inappropriately defasciculate along their pathway. The distribution of Sfrp1 and Sfrp2 in the eye is consistent with the idea that Sfrp1/2 normally constrain axon growth into the fiber layer and the optic disc. Disheveled axon growth instead seems linked to Sfrp-mediated modulation of metalloproteinase activity. Indeed, retinal explants from embryos with different Sfrp-null alleles or explants overexpressing ADAM10 extend axons with a disheveled appearance, which is reverted by the addition of Sfrp1 or an ADAM10-specific inhibitor. This mode of growth is associated with an abnormal proteolytic processing of L1 and N-cadherin, two ADAM10 substrates previously implicated in axon guidance. We thus propose that Sfrps contribute to coordinate visual axon growth with a dual mechanism: by directly signaling at the growth cone and by regulating the processing of other relevant cues.
Collapse
|
30
|
Miao W, Zhu B, Xiao X, Li Y, Dirbaba NB, Zhou B, Wu H. Effects of titanium dioxide nanoparticles on lead bioconcentration and toxicity on thyroid endocrine system and neuronal development in zebrafish larvae. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 161:117-126. [PMID: 25703175 DOI: 10.1016/j.aquatox.2015.02.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/30/2015] [Accepted: 02/02/2015] [Indexed: 06/04/2023]
Abstract
Nanoparticles (NPs) have attracted considerable attention because of their wide range of applications. Interactions between heavy metals (e.g., Pb) and NPs in aquatic environments may modify the bioavailability and toxicity of heavy metals. Therefore, this study investigated the influence of NPs (e.g., nano-TiO2) on the bioavailability and toxicity of Pb and its effects in the thyroid endocrine and nervous systems of zebrafish (Danio rerio) larvae. Zebrafish embryos (2-h post-fertilization) were exposed to five concentrations of Pb alone (0, 5, 10, 20, and 30μg/L) or in combination with nano-TiO2 (0.1mg/L) until 6 days post-fertilization. Results showed that the bioconcentration of Pb was significantly enhanced when combined with nano-TiO2 than when used alone. Zebrafish exposure to Pb alone at 30μg/L significantly decreased the thyroid hormone levels (T4 and T3), whereas nano-TiO2 treatment alone did not produce detectable changes. The levels of T4 and T3 were further decreased when Pb was combined with nano-TiO2 than when used alone. The transcription of the thyroid hormone-related factor tg gene was remarkably down-regulated by Pb treatment alone but up-regulated when Pb was combined with nano-TiO2. The significant up-regulation of tshβ gene and the down-regulation of TTR gene expression in the hypothalamic-pituitary-thyroid were observed in Pb with or without nano-TiO2 treatment groups. In addition, the transcription of genes involved in central nervous system (CNS) development (α-tubulin, mbp, gfap and shha) were significantly down-regulated by Pb and nano-TiO2 co-exposure as compared with Pb exposure alone. The locomotion activity analyzes confirmed that nano-TiO2 might enhance the toxicity of Pb to CNS development. These results suggest that nano-TiO2 increase bioconcentration of lead, which lead to the disruption of thyroid endocrine and neuronal system in zebrafish larvae.
Collapse
Affiliation(s)
- Wei Miao
- School of Environmental Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Biran Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; Graduate University of Chinese Academy of Sciences, Beijing 100039, China
| | - Xiaohong Xiao
- College of Mechanical and Electronic Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Ying Li
- School of Environmental Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Niguse Bekele Dirbaba
- School of Environmental Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Hongjuan Wu
- School of Environmental Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
31
|
El-Magd MA, Saleh AA, Farrag F, Abd El-Aziz RM, Ali HA, Salama MF. Regulation of Chick Ebf1-3 Gene Expression in the Pharyngeal Arches, Cranial Sensory Ganglia and Placodes. Cells Tissues Organs 2015; 199:278-93. [DOI: 10.1159/000369880] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2014] [Indexed: 11/19/2022] Open
|
32
|
El-Magd MA, Allen S, McGonnell I, Mansour AA, Otto A, Patel K. Shh regulates chick Ebf1 gene expression in somite development. Gene 2015; 554:87-95. [DOI: 10.1016/j.gene.2014.10.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 10/09/2014] [Accepted: 10/13/2014] [Indexed: 11/16/2022]
|
33
|
Abstract
The visual system is beautifully crafted to transmit information of the external world to visual processing and cognitive centers in the brain. For visual information to be relayed to the brain, a series of axon pathfinding events must take place to ensure that the axons of retinal ganglion cells, the only neuronal cell type in the retina that sends axons out of the retina, find their way out of the eye to connect with targets in the brain. In the past few decades, the power of molecular and genetic tools, including the generation of genetically manipulated mouse lines, have multiplied our knowledge about the molecular mechanisms involved in the sculpting of the visual system. Here, we review major advances in our understanding of the mechanisms controlling the differentiation of RGCs, guidance of their axons from the retina to the primary visual centers, and the refinement processes essential for the establishment of topographic maps and eye-specific axon segregation. Human disorders, such as albinism and achiasmia, that impair RGC axon growth and guidance and, thus, the establishment of a fully functioning visual system will also be discussed.
Collapse
Affiliation(s)
- Lynda Erskine
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Scotland, UK
| | - Eloisa Herrera
- Instituto de Neurosciencias de Alicante, CSIC-UMH, San Juan de Alicante, Spain
| |
Collapse
|
34
|
Powell S, Vinod A, Lemons ML. Isolation and culture of dissociated sensory neurons from chick embryos. J Vis Exp 2014:51991. [PMID: 25286047 DOI: 10.3791/51991] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neurons are multifaceted cells that carry information essential for a variety of functions including sensation, motor movement, learning, and memory. Studying neurons in vivo can be challenging due to their complexity, their varied and dynamic environments, and technical limitations. For these reasons, studying neurons in vitro can prove beneficial to unravel the complex mysteries of neurons. The well-defined nature of cell culture models provides detailed control over environmental conditions and variables. Here we describe how to isolate, dissociate, and culture primary neurons from chick embryos. This technique is rapid, inexpensive, and generates robustly growing sensory neurons. The procedure consistently produces cultures that are highly enriched for neurons and has very few non-neuronal cells (less than 5%). Primary neurons do not adhere well to untreated glass or tissue culture plastic, therefore detailed procedures to create two distinct, well-defined laminin-containing substrata for neuronal plating are described. Cultured neurons are highly amenable to multiple cellular and molecular techniques, including co-immunoprecipitation, live cell imagining, RNAi, and immunocytochemistry. Procedures for double immunocytochemistry on these cultured neurons have been optimized and described here.
Collapse
Affiliation(s)
- Sarah Powell
- Department of Natural Sciences, Assumption College
| | - Amrit Vinod
- Department of Natural Sciences, Assumption College
| | | |
Collapse
|
35
|
Avilés EC, Wilson NH, Stoeckli ET. Sonic hedgehog and Wnt: antagonists in morphogenesis but collaborators in axon guidance. Front Cell Neurosci 2013; 7:86. [PMID: 23772206 PMCID: PMC3677135 DOI: 10.3389/fncel.2013.00086] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 05/21/2013] [Indexed: 12/31/2022] Open
Abstract
As indicated by their name, morphogens were first identified for their role in the formation of tissues early in development. Secreted from a source, they spread through the tissue to form gradients by which they affect the differentiation of precursor cells in a concentration-dependent manner. In this context, the antagonistic roles of the morphogens of the Wnt family and Sonic hedgehog (Shh) in the specification of cell types along the dorso-ventral axis of the neural tube have been studied in detail. However, more recently, morphogens have been demonstrated to act well beyond the early stages of nervous system development, as additional roles of morphogen gradients in vertebrate neural circuit formation have been identified. Both Wnt and Shh affect neural circuit formation at several stages by their influence on neurite extension, axon pathfinding and synapse formation. In this review, we will summarize the mechanisms of morphogen function during axon guidance in the vertebrate nervous system.
Collapse
Affiliation(s)
- Evelyn C Avilés
- Institute of Molecular Life Sciences, University of Zurich Zurich, Switzerland
| | | | | |
Collapse
|
36
|
Briscoe J, Thérond PP. The mechanisms of Hedgehog signalling and its roles in development and disease. Nat Rev Mol Cell Biol 2013; 14:416-29. [DOI: 10.1038/nrm3598] [Citation(s) in RCA: 1212] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
37
|
Dudanova I, Klein R. Integration of guidance cues: parallel signaling and crosstalk. Trends Neurosci 2013; 36:295-304. [PMID: 23485451 DOI: 10.1016/j.tins.2013.01.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/25/2013] [Accepted: 01/28/2013] [Indexed: 11/28/2022]
Abstract
Growing axons are exposed to various guidance cues en route to their targets. Although many guidance molecules have been identified and their effects on axon behavior extensively studied, how axons react to combinations of signals remains largely unexplored. We review recent studies investigating the combined actions of guidance cues present at the same choice points. Two main scenarios are emerging from these studies: parallel signaling and crosstalk between guidance systems. In the first case, cues act in an additive manner, whereas in the second case the outcome is non-additive and differs from the sum of individual effects, suggesting more complex signal integration in the growth cone. Some of the molecular mechanisms underlying these interactions are beginning to be unraveled.
Collapse
Affiliation(s)
- Irina Dudanova
- Department Molecules-Signaling-Development, Max Planck Institute of Neurobiology, Martinsried D-82152, Germany.
| | | |
Collapse
|
38
|
Zhang Y, Zhao S, Dong W, He S, Wang H, Zhang L, Tang Y, Guo J, Guo S. Cloning and bioinformatical analysis of the N-terminus of the sonic hedgehog gene. Neural Regen Res 2013; 8:258-63. [PMID: 25206596 PMCID: PMC4107527 DOI: 10.3969/j.issn.1673-5374.2013.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/26/2012] [Indexed: 11/18/2022] Open
Abstract
The sonic hedgehog protein not only plays a key role in early embryonic development, but also has essential effects on the adult nervous system, including neural stem cell proliferation, differentiation, migration and neuronal axon guidance. The N-terminal fragment of sonic hedgehog is the key functional element in this process. Therefore, this study aimed to clone and analyze the N-terminal fragment of the sonic hedgehog gene. Total RNA was extracted from the notochord of a Sprague-Dawley rat at embryonic day 9 and the N-terminal fragment of sonic hedgehog was amplified by nested reverse transcription-PCR. The N-terminal fragment of the sonic hedgehog gene was successfully cloned. The secondary and tertiary structures of the N-terminal fragment of the sonic hedgehog protein were predicted using Jpred and Phyre online.
Collapse
Affiliation(s)
- Yi Zhang
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, China ; Department of Gynecology and Obstetrics, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong Province, China
| | - Shu Zhao
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Weiren Dong
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Suifen He
- Department of Laboratory, Shunde Guizhou Hospital, Foshan 528305, Guangdong Province, China
| | - Haihong Wang
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Lihua Zhang
- School of Medical Sciences, Foshan University, Foshan 528000, Guangdong Province, China
| | - Yinjuan Tang
- Department of Histology & Embryology, Xiangnan College, Chenzhou 418000, Hunan Province, China
| | - Jiasong Guo
- Department of Gynecology and Obstetrics, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong Province, China
| | - Suiqun Guo
- Department of Gynecology and Obstetrics, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong Province, China
| |
Collapse
|
39
|
Bijlsma MF, Damhofer H, Roelink H. Hedgehog-stimulated chemotaxis is mediated by smoothened located outside the primary cilium. Sci Signal 2012; 5:ra60. [PMID: 22912493 DOI: 10.1126/scisignal.2002798] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Regulation of the Hedgehog (Hh) pathway relies on an interaction of two receptors. In the absence of Hh, Patched1 (Ptch1) inhibits the pathway. Binding of the ligand Hh to Ptch1 stimulates the localization of the activating receptor Smoothened (Smo) to the primary cilium, which is required for the transcriptional Hh response. Hh can also induce chemotaxis through a nontranscriptional pathway. We assessed the effects of defective ciliary localization of Smo on its subcellular trafficking and ability to mediate chemotactic signaling. Cells expressing mutants of Smo that could not localize to the primary cilium or cells lacking the primary cilium showed altered intracellular trafficking of Smo and, in response to Hh or Smo agonists, decreased transcriptional signaling and enhanced chemotactic responsiveness. Thus, the ciliary localization machinery appears to transport Smo to subcellular sites where it can mediate transcriptional signaling and away from locations where it can mediate chemotactic signaling. The subcellular localization of Smo is thus a crucial determinant of its signaling characteristics and implies the existence of a pool of Smo dedicated to chemotaxis.
Collapse
Affiliation(s)
- Maarten F Bijlsma
- Department of Molecular and Cell Biology, University of California, Berkeley, 16 Barker Hall, 3204, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
40
|
Stacher Hörndli C, Chien CB. Sonic hedgehog is indirectly required for intraretinal axon pathfinding by regulating chemokine expression in the optic stalk. Development 2012; 139:2604-13. [PMID: 22696293 DOI: 10.1242/dev.077594] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Successful axon pathfinding requires both correct patterning of tissues, which will later harbor axonal tracts, and precise localization of axon guidance cues along these tracts at the time of axon outgrowth. Retinal ganglion cell (RGC) axons grow towards the optic disc in the central retina, where they turn to exit the eye through the optic nerve. Normal patterning of the optic disc and stalk and the expression of guidance cues at this choice point are necessary for the exit of RGC axons out of the eye. Sonic hedgehog (Shh) has been implicated in both patterning of ocular tissue and direct guidance of RGC axons. Here, we examine the precise spatial and temporal requirement for Hedgehog (Hh) signaling for intraretinal axon pathfinding and show that Shh acts to pattern the optic stalk in zebrafish but does not guide RGC axons inside the eye directly. We further reveal an interaction between the Hh and chemokine pathways for axon guidance and show that cxcl12a functions downstream of Shh and depends on Shh for its expression at the optic disc. Together, our results support a model in which Shh acts in RGC axon pathfinding indirectly by regulating axon guidance cues at the optic disc through patterning of the optic stalk.
Collapse
Affiliation(s)
- Cornelia Stacher Hörndli
- Program in Neuroscience, Department of Neurobiology and Anatomy, University of Utah Medical Center, 20 North 1900 East, Salt Lake City, UT 84132, USA.
| | | |
Collapse
|
41
|
Guo D, Standley C, Bellve K, Fogarty K, Bao ZZ. Protein kinase Cα and integrin-linked kinase mediate the negative axon guidance effects of Sonic hedgehog. Mol Cell Neurosci 2012; 50:82-92. [PMID: 22521536 PMCID: PMC3383945 DOI: 10.1016/j.mcn.2012.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Revised: 03/22/2012] [Accepted: 03/26/2012] [Indexed: 01/22/2023] Open
Abstract
In addition to its role as a morphogen, Sonic hedgehog (Shh) has also been shown to function as a guidance factor that directly acts on the growth cones of various types of axons. However, the noncanonical signaling pathways that mediate the guidance effects of Shh protein remain poorly understood. We demonstrate that a novel signaling pathway consisting of protein kinase Cα (PKCα) and integrin-linked kinase (ILK) mediates the negative guidance effects of high concentration of Shh on retinal ganglion cell (RGC) axons. Shh rapidly increased Ca(2+) level and activated PKCα and ILK in the growth cones of RGC axons. By in vitro kinase assay, PKCα was found to directly phosphorylate ILK on threonine-173 and -181. Inhibition of PKCα or expression of a mutant ILK with the PKCα phosphorylation sites mutated (ILK-DM), abolished the Shh-induced macropinocytosis, growth cone collapse and repulsive axon turning. In vivo, expression of a dominant negative PKCα or ILK-DM disrupted RGC axon pathfinding at the optic chiasm but not the projection toward the optic disk, supporting that this signaling pathway plays a specific role in Shh-mediated negative guidance effects.
Collapse
Affiliation(s)
- Daorong Guo
- Department of Medicine and Cell Biology, Program in Neuroscience, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | | | | | | | | |
Collapse
|
42
|
Jung IH, Jung DE, Park YN, Song SY, Park SW. Aberrant Hedgehog ligands induce progressive pancreatic fibrosis by paracrine activation of myofibroblasts and ductular cells in transgenic zebrafish. PLoS One 2011; 6:e27941. [PMID: 22164219 PMCID: PMC3229500 DOI: 10.1371/journal.pone.0027941] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 10/28/2011] [Indexed: 01/11/2023] Open
Abstract
Hedgehog (Hh) signaling is frequently up-regulated in fibrogenic pancreatic diseases including chronic pancreatitis and pancreatic cancer. Although recent series suggest exclusive paracrine activation of stromal cells by Hh ligands from epithelial components, debates still exist on how Hh signaling works in pathologic conditions. To explore how Hh signaling affects the pancreas, we investigated transgenic phenotypes in zebrafish that over-express either Indian Hh or Sonic Hh along with green fluorescence protein (GFP) to enable real-time observation, or GFP alone as control, at the ptf1a domain. Transgenic embryos and zebrafish were serially followed for transgenic phenotypes, and investigated using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), in situ hybridization, and immunohistochemistry. Over-expression of Ihh or Shh reveals virtually identical phenotypes. Hh induces morphologic changes in a developing pancreas without derangement in acinar differentiation. In older zebrafish, Hh induces progressive pancreatic fibrosis intermingled with proliferating ductular structures, which is accompanied by the destruction of the acinar structures. Both myofibroblasts and ductular are activated and proliferated by paracrine Hh signaling, showing restricted expression of Hh downstream components including Patched1 (Ptc1), Smoothened (Smo), and Gli1/2 in those Hh-responsive cells. Hh ligands induce matrix metalloproteinases (MMPs), especially MMP9 in all Hh-responsive cells, and transform growth factor-ß1 (TGFß1) only in ductular cells. Aberrant Hh over-expression, however, does not induce pancreatic tumors. On treatment with inhibitors, embryonic phenotypes are reversed by either cyclopamine or Hedgehog Primary Inhibitor-4 (HPI-4). Pancreatic fibrosis is only prevented by HPI-4. Our study provides strong evidence of Hh signaling which induces pancreatic fibrosis through paracrine activation of Hh-responsive cells in vivo. Induction of MMPs and TGFß1 by Hh signaling expands on the current understanding of how Hh signaling affects fibrosis and tumorigenesis. These transgenic models will be a valuable platform in exploring the mechanism of fibrogenic pancreatic diseases which are induced by Hh signaling activation.
Collapse
Affiliation(s)
- In Hye Jung
- Postgraduate School of National Core Research Center for Nanomedical Technology, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dawoon E. Jung
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Nyun Park
- Department of Pathology, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Si Young Song
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Woo Park
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
43
|
Shh signaling guides spatial pathfinding of raphespinal tract axons by multidirectional repulsion. Cell Res 2011; 22:697-716. [PMID: 22064698 DOI: 10.1038/cr.2011.172] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Relatively little is known about the molecular mechanisms underlying spatial pathfinding in the descending serotonergic raphespinal tract (RST) in the developing spinal cord, one of the most important nerve pathways for pain, sensory and motor functions. We provide evidence that ventral floor plate-secreted Sonic hedgehog (Shh) is responsible for the establishment of decreasing gradients in both the anterior-to-posterior (A-P) and the medial-to-lateral (M-L) directions in the ventral spinal cord during serotonergic RST axon projection. Downstream components of the Shh pathway, Patched 1 (Ptch1) and Smoothened (Smo), were expressed in the serotonergic caudal raphe nuclei and enriched in the descending serotonergic RST axons. Diffusible Shh repulsion of serotonergic RST axons was shown to be mediated by Shh-Ptch1 interactions and derepression of Smo. Using a co-culture assay, we showed that A-P graded repulsion mediated by Shh signaling pushed the serotonergic axons caudally through the ventral spinal cord and M-L graded repulsion mediated by Shh signaling simultaneously restricted the serotonergic axons to the ventral and ventral-lateral funiculus. Prominent pathfinding errors of serotonergic RST axons were observed in various Shh, Ptch1 and Smo mutants. We conclude that Shh signaling-mediated multidirectional repulsion is required to push descending serotonergic RST axons in the A-P direction, and to restrict these axons to the ventral and ventral-lateral funiculus in the M-L direction. This is the first demonstration that Shh signaling-mediated multidirectional repulsion of serotonergic RST axons maintains spatial axon pathfinding in the developing spinal cord.
Collapse
|
44
|
Atkinson-Leadbeater K, McFarlane S. Extrinsic factors as multifunctional regulators of retinal ganglion cell morphogenesis. Dev Neurobiol 2011; 71:1170-85. [DOI: 10.1002/dneu.20924] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
45
|
Zhang J, Peterson SM, Weber GJ, Zhu X, Zheng W, Freeman JL. Decreased axonal density and altered expression profiles of axonal guidance genes underlying lead (Pb) neurodevelopmental toxicity at early embryonic stages in the zebrafish. Neurotoxicol Teratol 2011; 33:715-20. [PMID: 21839828 DOI: 10.1016/j.ntt.2011.07.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 07/26/2011] [Accepted: 07/28/2011] [Indexed: 11/17/2022]
Abstract
Previous studies have reported that environmental lead (Pb) exposure can result in neurological alterations in children leading to reduced IQ, attention deficit hyperactivity disorder, and diminished reading and learning abilities. However, the specific alterations in neurodevelopmental morphology and the underlying genetic mechanisms of these alterations have not yet been thoroughly defined. To investigate alterations in neurologic morphology and test the hypothesis that developmental Pb neurotoxicity is partially mediated through alterations in neuronal growth and transport function of axons, the changes of specific axon tracts in the embryonic zebrafish brain were observed with anti-acetylated α-tubulin staining at several developmental time points through 36hours post fertilization (hpf). In addition, the role of a subset of axonogenesis-related genes including shha, epha4b, netrin1b, netrin2, and noiwas investigated with real-time quantitative PCR (qPCR). Pb treatment resulted in decreased axonal density at 18, 20, and 24hpf for specific axon tracts in the midbrain and forebrain. These observations corresponded to an observed down-regulation of shha and epha4b at 14 and 16hpf, respectively. The axonal density in Pb exposed individuals at later stages (30 and 36hpf) was not significantly different from controls. An overexpression of netrin2 at these two developmental stages suggests a novel role for this gene in regulating axonal density specific to Pb neurotoxicity. Although no significant differences in axonal density was observed in the two later developmental stages, further studies are needed to determine if the morphologic alterations observed at the earlier stages will have lasting functional impacts.
Collapse
Affiliation(s)
- Jun Zhang
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | | | | | | | | | | |
Collapse
|
46
|
Kunzevitzky NJ, Almeida MV, Duan Y, Li S, Xiang M, Goldberg JL. Foxn4 is required for retinal ganglion cell distal axon patterning. Mol Cell Neurosci 2011; 46:731-41. [PMID: 21334440 DOI: 10.1016/j.mcn.2011.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 01/10/2011] [Accepted: 02/10/2011] [Indexed: 10/18/2022] Open
Abstract
The regulation of retinal ganglion cell (RGC) axon growth and patterning in vivo is thought to be largely dependent on interactions with visual pathway and target cells. Here we address the hypothesis that amacrine cells, RGCs' presynaptic partners, regulate RGC axon growth or targeting. We asked whether amacrine cells play a role in RGC axon growth in vivo using Foxn4(-/-) mice, which have fewer amacrine cells, but a normal complement of RGCs. We found that Foxn4(-/-) mice have a similar reduction in most subtypes of amacrine cells examined. Remarkably, spontaneous retinal waves were not affected by the reduction of amacrine cells in the Foxn4(-/-) mice. There was, however, a developmental delay in the distribution of RGC projections to the superior colliculus. Furthermore, RGC axons failed to penetrate into the retinorecipient layers in the Foxn4(-/-) mice. Foxn4 is not expressed by RGCs and was not detectable in the superior colliculus itself. These findings suggest that amacrine cells are critical for proper RGC axon growth in vivo, and support the hypothesis that the amacrine cell-RGC interaction may contribute to the regulation of distal projections and axon patterning.
Collapse
Affiliation(s)
- Noelia J Kunzevitzky
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | |
Collapse
|
47
|
Gordon L, Mansh M, Kinsman H, Morris AR. Xenopus sonic hedgehog guides retinal axons along the optic tract. Dev Dyn 2011; 239:2921-32. [PMID: 20931659 DOI: 10.1002/dvdy.22430] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The role of classic morphogens such as Sonic hedgehog (Shh) as axon guidance cues has been reported in a variety of vertebrate organisms (Charron and Tessier-Lavigne [2005] Development 132:2251-2262). In this work, we provide the first evidence that Xenopus sonic hedgehog (Xshh) signaling is involved in guiding retinal ganglion cell (RGC) axons along the optic tract. Xshh is expressed in the brain during retinal axon extension, adjacent to these axons in the ventral diencephalon. Retinal axons themselves express Patched 1 and Smoothened co-receptors during RGC axon growth. Blocking Shh signaling causes abnormal ventral pathfinding, and targeting errors at the optic tectum. Misexpression of exogenous N-Shh peptide in vivo also causes pathfinding errors. Retinal axons grown in culture respond to N-Shh in a dose-dependent manner, either by decreasing extension at lower concentrations, or retracting axons in the presence of higher doses. These data suggest that Shh signaling is required for normal RGC axon pathfinding and tectal targeting in the developing visual system of Xenopus. We propose that Shh serves as a ventral optic tract repellent that helps to define the caudal boundary for retinal axons in the diencephalon, and that this signaling is also required for initial target recognition at the optic tectum.
Collapse
Affiliation(s)
- Laura Gordon
- Haverford College Department of Biology, Haverford, Pennsylvania 19041, USA
| | | | | | | |
Collapse
|
48
|
Angeloni NL, Bond CW, Tang Y, Harrington DA, Zhang S, Stupp SI, McKenna KE, Podlasek CA. Regeneration of the cavernous nerve by Sonic hedgehog using aligned peptide amphiphile nanofibers. Biomaterials 2010; 32:1091-101. [PMID: 20971506 DOI: 10.1016/j.biomaterials.2010.10.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 10/01/2010] [Indexed: 02/07/2023]
Abstract
SHH plays a significant role in peripheral nerve regeneration and has clinical potential to be used as a regenerative therapy for the CN in prostatectomy patients and in other patients with neuropathy of peripheral nerves. Efforts to regenerate the cavernous nerve (CN), which provides innervation to the penis, have been minimally successful, with little translation into improved clinical outcomes. We propose that, Sonic hedgehog (SHH), is critical to maintain CN integrity, and that SHH delivered to the CN by novel peptide amphiphile (PA) nanofibers, will promote CN regeneration, restore physiological function, and prevent penile morphology changes that result in erectile dysfunction (ED). We performed localization studies, inhibition of SHH signaling in the CN, and treatment of crushed CNs with SHH protein via linear PA gels, which are an innovative extended release method of delivery. Morphological, functional and molecular analysis revealed that SHH protein is essential to maintain CN architecture, and that SHH treatment promoted CN regeneration, suppressed penile apoptosis and caused a 58% improvement in erectile function in less than half the time reported in the literature. These studies show that SHH has substantial clinical application to regenerate the CN in prostatectomy and diabetic patients, that this methodology has broad application to regenerate any peripheral nerve that SHH is necessary for maintenance of its structure, and that this nanotechnology method of protein delivery may have wide spread application as an in vivo delivery tool in many organs.
Collapse
Affiliation(s)
- Nicholas L Angeloni
- Department of Urology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Fiorini Z, Jasoni CL. A novel developmental role for kisspeptin in the growth of gonadotrophin-releasing hormone neurites to the median eminence in the mouse. J Neuroendocrinol 2010; 22:1113-25. [PMID: 20722977 DOI: 10.1111/j.1365-2826.2010.02059.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The puberty- and fertility-regulating neuropeptide kisspeptin (KISS1) exerts dramatic effects on the physiology of adult gonadotrophin-releasing hormone (GnRH) neurones as a master regulator of mammalian reproduction. Given the action of KISS1 directly on adult GnRH neurones, and that KISS1 activates a signal transduction cascade involved in neurite growth in other neurones, we investigated whether KISS1 may play a role in the normal growth of GnRH neurites to the median eminence. A reverse transcription-polymerase chain reaction demonstrated the expression of Kiss1 mRNA in the embryonic mediobasal hypothalamus, the target region for GnRH neurite termination, as early as embryonic day 13.5 (E13.5), a time when the first GnRH neurites are arriving. Complementary expression of the mRNA encoding the KISS1 receptor, Kiss1r, in the preoptic area (POA) at E13.5 was also observed, suggesting that POA-resident GnRH neurones can respond to KISS1 from an early age. To examine the effects of KISS1 on GnRH neurite growth in isolation, E15.5 POA explants, containing GnRH neurones actively extending neurites, were grown in three-dimensional collagen gels. In the presence of KISS1 (1 μm), both the number and length of GnRH neurites were increased significantly compared to controls without KISS1. The effects of KISS1 on GnRH neurite growth could be inhibited by pretreatment with the phospholipase C inhibitor U73122 (50 μm), indicating that embryonic and adult GnRH neurones respond to KISS1 with the same intracellular signalling pathway. KISS1 provided in a concentration gradient from a fixed source had no effect on GnRH neurite growth, indicating that KISS1 does not function as a long-range chemoattractant. Taken together, these results identify KISS1 as a stimulator of GnRH neurite growth, and suggest that it influences GnRH neurites at close-range to innervate the median eminence. These data add a novel developmental role to the repertoire of the functions of KISS1 in mammalian reproduction.
Collapse
Affiliation(s)
- Z Fiorini
- Centre for Neuroendocrinology, Department of Anatomy & Structural Biology, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | | |
Collapse
|
50
|
Cooper DL, Gentle JE, Barreto E, Olds JL. Synchronized changes to relative neuron populations in postnatal human neocortical development. Cogn Neurodyn 2010; 4:151-63. [PMID: 21629587 PMCID: PMC2866365 DOI: 10.1007/s11571-010-9103-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 01/15/2010] [Indexed: 01/19/2023] Open
Abstract
UNLABELLED Mammalian prenatal neocortical development is dominated by the synchronized formation of the laminae and migration of neurons. Postnatal development likewise contains "sensitive periods" during which functions such as ocular dominance emerge. Here we introduce a novel neuroinformatics approach to identify and study these periods of active development. Although many aspects of the approach can be used in other studies, some specific techniques were chosen because of a legacy dataset of human histological data (Conel in The postnatal development of the human cerebral cortex, vol 1-8. Harvard University Press, Cambridge, 1939-1967). Our method calculates normalized change vectors from the raw histological data, and then employs k-means cluster analysis of the change vectors to explore the population dynamics of neurons from 37 neocortical areas across eight postnatal developmental stages from birth to 72 months in 54 subjects. We show that the cortical "address" (Brodmann area/sub-area and layer) provides the necessary resolution to segregate neuron population changes into seven correlated "k-clusters" in k-means cluster analysis. The members in each k-cluster share a single change interval where the relative share of the cortex by the members undergoes its maximum change. The maximum change occurs in a different change interval for each k-cluster. Each k-cluster has at least one totally connected maximal "clique" which appears to correspond to cortical function. ELECTRONIC SUPPLEMENTARY MATERIAL The online version of this article (doi:10.1007/s11571-010-9103-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David L. Cooper
- Doctoral Program in Neuroscience, George Mason University, Fairfax, VA 22030 USA
| | - James E. Gentle
- Department of Computational and Data Sciences, George Mason University, Fairfax, VA 22030 USA
| | - Ernest Barreto
- Department of Physics & Astronomy, the Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030 USA
| | - James L. Olds
- Department of Molecular Neuroscience, Krasnow Institute for Advanced Study, George Mason University, 4400 University Dr., Fairfax, VA 22030 USA
| |
Collapse
|