1
|
Hlavac K, Pavelkova P, Ondrisova L, Mraz M. FoxO1 signaling in B cell malignancies and its therapeutic targeting. FEBS Lett 2024. [PMID: 39533662 DOI: 10.1002/1873-3468.15057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
FoxO transcription factors (FoxO1, FoxO3a, FoxO4, FoxO6) are a highly evolutionary conserved subfamily of the 'forkhead' box proteins. They have traditionally been considered tumor suppressors, but FoxO1 also exhibits oncogenic properties. The complex nature of FoxO1 is illustrated by its various roles in B cell development and differentiation, immunoglobulin gene rearrangement and cell-surface B cell receptor (BCR) structure, DNA damage control, cell cycle regulation, and germinal center reaction. FoxO1 is tightly regulated at a transcriptional (STAT3, HEB, EBF, FoxOs) and post-transcriptional level (Akt, AMPK, CDK2, GSK3, IKKs, JNK, MAPK/Erk, SGK1, miRNA). In B cell malignancies, recurrent FoxO1 activating mutations (S22/T24) and aberrant nuclear export and activity have been described, underscoring the potential of its therapeutic inhibition. Here, we review FoxO1's roles across B cell and myeloid malignancies, namely acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), diffuse large B cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Burkitt lymphoma (BL), Hodgkin lymphoma (HL), and multiple myeloma (MM). We also discuss preclinical evidence for FoxO1 targeting by currently available inhibitors (AS1708727, AS1842856, cpd10).
Collapse
Affiliation(s)
- Krystof Hlavac
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Petra Pavelkova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Laura Ondrisova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Marek Mraz
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| |
Collapse
|
2
|
Giunti S, Blanco MG, De Rosa MJ, Rayes D. The ketone body β-hydroxybutyrate ameliorates neurodevelopmental deficits in the GABAergic system of daf-18/PTEN Caenorhabditis elegans mutants. eLife 2024; 13:RP94520. [PMID: 39422188 PMCID: PMC11488850 DOI: 10.7554/elife.94520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
A finely tuned balance between excitation and inhibition (E/I) is essential for proper brain function. Disruptions in the GABAergic system, which alter this equilibrium, are a common feature in various types of neurological disorders, including autism spectrum disorders (ASDs). Mutations in Phosphatase and Tensin Homolog (PTEN), the main negative regulator of the phosphatidylinositol 3-phosphate kinase/Akt pathway, are strongly associated with ASD. However, it is unclear whether PTEN deficiencies can differentially affect inhibitory and excitatory signaling. Using the Caenorhabditis elegans neuromuscular system, where both excitatory (cholinergic) and inhibitory (GABAergic) inputs regulate muscle activity, we found that daf-18/PTEN mutations impact GABAergic (but not cholinergic) neurodevelopment and function. This selective impact results in a deficiency in inhibitory signaling. The defects observed in the GABAergic system in daf-18/PTEN mutants are due to reduced activity of DAF-16/FOXO during development. Ketogenic diets (KGDs) have proven effective for disorders associated with E/I imbalances. However, the mechanisms underlying their action remain largely elusive. We found that a diet enriched with the ketone body β-hydroxybutyrate during early development induces DAF-16/FOXO activity, therefore improving GABAergic neurodevelopment and function in daf-18/PTEN mutants. Our study provides valuable insights into the link between PTEN mutations and neurodevelopmental defects and delves into the mechanisms underlying the potential therapeutic effects of KGDs.
Collapse
Affiliation(s)
- Sebastián Giunti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| | - María Gabriela Blanco
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| | - María José De Rosa
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| | - Diego Rayes
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| |
Collapse
|
3
|
Romussi S, Giunti S, Andersen N, De Rosa MJ. C. elegans: a prominent platform for modeling and drug screening in neurological disorders. Expert Opin Drug Discov 2024; 19:565-585. [PMID: 38509691 DOI: 10.1080/17460441.2024.2329103] [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: 11/13/2023] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Human neurodevelopmental and neurodegenerative diseases (NDevDs and NDegDs, respectively) encompass a broad spectrum of disorders affecting the nervous system with an increasing incidence. In this context, the nematode C. elegans, has emerged as a benchmark model for biological research, especially in the field of neuroscience. AREAS COVERED The authors highlight the numerous advantages of this tiny worm as a model for exploring nervous system pathologies and as a platform for drug discovery. There is a particular focus given to describing the existing models of C. elegans for the study of NDevDs and NDegDs. Specifically, the authors underscore their strong applicability in preclinical drug development. Furthermore, they place particular emphasis on detailing the common techniques employed to explore the nervous system in both healthy and diseased states. EXPERT OPINION Drug discovery constitutes a long and expensive process. The incorporation of invertebrate models, such as C. elegans, stands as an exemplary strategy for mitigating costs and expediting timelines. The utilization of C. elegans as a platform to replicate nervous system pathologies and conduct high-throughput automated assays in the initial phases of drug discovery is pivotal for rendering therapeutic options more attainable and cost-effective.
Collapse
Affiliation(s)
- Stefano Romussi
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
| | - Sebastián Giunti
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Natalia Andersen
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - María José De Rosa
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| |
Collapse
|
4
|
Suryawinata N, Yokosawa R, Tan KHC, Lai AL, Sone R, Mori I, Noma K. Dietary E. coli promotes age-dependent chemotaxis decline in C. elegans. Sci Rep 2024; 14:5529. [PMID: 38448519 PMCID: PMC10918063 DOI: 10.1038/s41598-024-52272-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/16/2024] [Indexed: 03/08/2024] Open
Abstract
An animal's ability to sense odors declines during aging, and its olfactory drive is tuned by internal states such as satiety. However, whether internal states modulate an age-dependent decline in odor sensation is unknown. To address this issue, we utilized the nematode Caenorhabditis elegans and compared their chemotaxis abilities toward attractive odorants when aged under different dietary conditions. Feeding with the standard laboratory diet, Escherichia coli attenuated the chemotaxis ability toward diacetyl, isoamyl alcohol, and benzaldehyde when aged. On the other hand, feeding with either the lactic acid bacteria Lactobacillus reuteri or food deprivation selectively maintained the chemotaxis ability toward diacetyl. Our results suggest that ingestion of E. coli causes age-dependent chemotaxis decline. The changes in the chemotaxis behavior are attributed to the different expressions of diacetyl receptor odr-10, and the chemotaxis behavior of aged animals under food deprivation is shown to be dependent on daf-16. Our study demonstrates the molecular mechanism of how diet shapes the trajectory of age-dependent decline in chemosensory behaviors.
Collapse
Affiliation(s)
- Nadia Suryawinata
- Group of Nutritional Neuroscience, Graduate School of Science, Neuroscience Institute, Nagoya University, Nagoya, 464-8602, Japan
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Blvd, Pasadena, CA, 91125, USA
| | - Rikuou Yokosawa
- Group of Nutritional Neuroscience, Graduate School of Science, Neuroscience Institute, Nagoya University, Nagoya, 464-8602, Japan
- Group of Microbial Motility, Division of Natural Science, Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, 464-8602, Japan
| | - Ke Hui Cassandra Tan
- Group of Nutritional Neuroscience, Graduate School of Science, Neuroscience Institute, Nagoya University, Nagoya, 464-8602, Japan
| | - Alison Lok Lai
- Group of Nutritional Neuroscience, Graduate School of Science, Neuroscience Institute, Nagoya University, Nagoya, 464-8602, Japan
| | - Ryusei Sone
- Group of Nutritional Neuroscience, Graduate School of Science, Neuroscience Institute, Nagoya University, Nagoya, 464-8602, Japan
- Group of Microbial Motility, Division of Natural Science, Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, 464-8602, Japan
| | - Ikue Mori
- Group of Molecular Neurobiology, Graduate School of Science, Neuroscience Institute, Nagoya University, Nagoya, 464-8602, Japan
| | - Kentaro Noma
- Group of Nutritional Neuroscience, Graduate School of Science, Neuroscience Institute, Nagoya University, Nagoya, 464-8602, Japan.
- Group of Microbial Motility, Division of Natural Science, Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, 464-8602, Japan.
| |
Collapse
|
5
|
Higurashi S, Tsukada S, Aleogho BM, Park JH, Al-Hebri Y, Tanaka M, Nakano S, Mori I, Noma K. Bacterial diet affects the age-dependent decline of associative learning in Caenorhabditis elegans. eLife 2023; 12:81418. [PMID: 37252859 DOI: 10.7554/elife.81418] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
The causality and mechanism of dietary effects on brain aging are still unclear due to the long time scales of aging. The nematode Caenorhabditis elegans has contributed to aging research because of its short lifespan and easy genetic manipulation. When fed the standard laboratory diet, Escherichia coli, C. elegans experiences an age-dependent decline in temperature-food associative learning, called thermotaxis. To address if diet affects this decline, we screened 35 lactic acid bacteria as alternative diet and found that animals maintained high thermotaxis ability when fed a clade of Lactobacilli enriched with heterofermentative bacteria. Among them, Lactobacillus reuteri maintained the thermotaxis of aged animals without affecting their lifespan and motility. The effect of Lb. reuteri depends on the DAF-16 transcription factor functioning in neurons. Furthermore, RNA sequencing analysis revealed that differentially expressed genes between aged animals fed different bacteria were enriched with DAF-16 targets. Our results demonstrate that diet can impact brain aging in a daf-16-dependent manner without changing the lifespan.
Collapse
Affiliation(s)
- Satoshi Higurashi
- Milk Science Research Institute, Megmilk Snow Brand Co. Ltd., Saitama, Japan
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Sachio Tsukada
- Milk Science Research Institute, Megmilk Snow Brand Co. Ltd., Saitama, Japan
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Binta Maria Aleogho
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Group of Microbial Motility, Department of Biological Science, Division of Natural Science, Graduate school of Science, Nagoya University, Nagoya, Japan
| | - Joo Hyun Park
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yana Al-Hebri
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Masaru Tanaka
- Milk Science Research Institute, Megmilk Snow Brand Co. Ltd., Saitama, Japan
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Shunji Nakano
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Ikue Mori
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Kentaro Noma
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Group of Microbial Motility, Department of Biological Science, Division of Natural Science, Graduate school of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
6
|
Vogt MC, Hobert O. Starvation-induced changes in somatic insulin/IGF-1R signaling drive metabolic programming across generations. SCIENCE ADVANCES 2023; 9:eade1817. [PMID: 37027477 PMCID: PMC10081852 DOI: 10.1126/sciadv.ade1817] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 03/08/2023] [Indexed: 05/30/2023]
Abstract
Exposure to adverse nutritional and metabolic environments during critical periods of development can exert long-lasting effects on health outcomes of an individual and its descendants. Although such metabolic programming has been observed in multiple species and in response to distinct nutritional stressors, conclusive insights into signaling pathways and mechanisms responsible for initiating, mediating, and manifesting changes to metabolism and behavior across generations remain scarce. By using a starvation paradigm in Caenorhabditis elegans, we show that starvation-induced changes in dauer formation-16/forkhead box transcription factor class O (DAF-16/FoxO) activity, the main downstream target of insulin/insulin-like growth factor 1 (IGF-1) receptor signaling, are responsible for metabolic programming phenotypes. Tissue-specific depletion of DAF-16/FoxO during distinct developmental time points demonstrates that DAF-16/FoxO acts in somatic tissues, but not directly in the germline, to both initiate and manifest metabolic programming. In conclusion, our study deciphers multifaceted and critical roles of highly conserved insulin/IGF-1 receptor signaling in determining health outcomes and behavior across generations.
Collapse
|
7
|
Cuko L, Cale AR, Rambo L, Knoblock ML, Karp X. Distinct daf-16 isoforms regulate specification of vulval precursor cells in Caenorhabditis elegans. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000706. [PMID: 36575736 PMCID: PMC9790082 DOI: 10.17912/micropub.biology.000706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/29/2022]
Abstract
FOXO transcription factors regulate development, longevity, and stress-resistance across species. The C. elegans FOXO ortholog, daf-16, has three major isoforms with distinct promoters and N-termini. Different combinations of isoforms regulate different processes. Adverse environments can induce dauer diapause after the second larval molt. During dauer, daf-16 blocks specification of vulval precursor cells, including EGFR/Ras-mediated 1˚ fate specification and LIN-12/Notch-mediated 2˚ fate specification. Using isoform-specific mutants, we find that daf-16a and daf-16f are functionally redundant for the block to the expression of 1˚ fate markers. In contrast, all three isoforms contribute to blocking the expression of 2˚ fate markers.
Collapse
Affiliation(s)
| | - Allison R Cale
- Central Michigan University
,
Current address: University of Michigan, Ann Arbor
| | - Loni Rambo
- Central Michigan University
,
Current address: University of Michigan, Ann Arbor
| | | | - Xantha Karp
- Central Michigan University
,
Correspondence to: Xantha Karp (
)
| |
Collapse
|
8
|
Zhang Y, Zhou Q, Lu L, Zhao C, Zhang H, Liu R, Pu Y, Yin L. Integrating Transcriptomics and Free Fatty Acid Profiling Analysis Reveal Cu Induces Shortened Lifespan and Increased Fat Accumulation and Oxidative Damage in C. elegans. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5297342. [PMID: 36017239 PMCID: PMC9398846 DOI: 10.1155/2022/5297342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/03/2022] [Accepted: 08/01/2022] [Indexed: 11/28/2022]
Abstract
Nowadays, human beings are exposed to Cu in varieties of environmental mediums, resulting in health risks needing urgent attention. Our research found that Cu shortened lifespan and induced aging-related phenotypes of Caenorhabditis elegans (C. elegans). Transcriptomics data showed differential expression genes induced by Cu were mainly involved in regulation of metabolism and longevity, especially in fatty acid metabolism. Quantitative detection of free fatty acid by GC/MS further found that Cu upregulated free fatty acids of C. elegans. A mechanism study confirmed that Cu promoted the fat accumulation in nematodes, which was owing to disorder of fatty acid desaturase and CoA synthetase, endoplasmic reticulum unfolded protein response (UPRER), mitochondrial membrane potential, and unfolded protein response (UPRmt). In addition, Cu activated oxidative stress and prevented DAF-16 translocating into nuclear with a concomitant reduction in the expression of environmental stress-related genes. Taken together, the research suggested that Cu promoted aging and induced fat deposition and oxidative damage.
Collapse
Affiliation(s)
- Ying Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China; School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Qian Zhou
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China; School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Lu Lu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China; School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Chao Zhao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China; School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Hu Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China; School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China; School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China; School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China; School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
9
|
Jofré DM, Hoffman DK, Cervino AS, Hahn GM, Grundy M, Yun S, Amrit FRG, Stolz DB, Godoy LF, Salvatore E, Rossi FA, Ghazi A, Cirio MC, Yanowitz JL, Hochbaum D. The CHARGE syndrome ortholog CHD-7 regulates TGF-β pathways in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2022; 119:e2109508119. [PMID: 35394881 PMCID: PMC9169646 DOI: 10.1073/pnas.2109508119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 02/24/2022] [Indexed: 11/18/2022] Open
Abstract
CHARGE syndrome is a complex developmental disorder caused by mutations in the chromodomain helicase DNA-binding protein-7 (CHD7) and characterized by retarded growth and malformations in the heart and nervous system. Despite the public health relevance of this disorder, relevant cellular pathways and targets of CHD7 that relate to disease pathology are still poorly understood. Here we report that chd-7, the nematode ortholog of Chd7, is required for dauer morphogenesis, lifespan determination, stress response, and body size determination. Consistent with our discoveries, we found chd-7 to be allelic to scd-3, a previously identified dauer suppressor from the DAF-7/ tumor growth factor-β (TGF-β) pathway. Epistatic analysis places CHD-7 at the level of the DAF-3/DAF-5 complex, but we found that CHD-7 also directly impacts the expression of multiple components of this pathway. Transcriptomic analysis revealed that chd-7 mutants fail to repress daf-9 for execution of the dauer program. In addition, CHD-7 regulates the DBL-1/BMP pathway components and shares roles in male tail development and cuticle synthesis. To explore a potential conserved function for chd-7 in vertebrates, we used Xenopus laevis embryos, an established model to study craniofacial development. Morpholino-mediated knockdown of Chd7 led to a reduction in col2a1 messenger RNA (mRNA) levels, a collagen whose expression depends on TGF-β signaling. Both embryonic lethality and craniofacial defects in Chd7-depleted tadpoles were partially rescued by overexpression of col2a1 mRNA. We suggest that Chd7 has conserved roles in regulation of the TGF-β signaling pathway and pathogenic Chd7 could lead to a defective extracellular matrix deposition.
Collapse
Affiliation(s)
- Diego M. Jofré
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| | | | - Ailen S. Cervino
- Instituto de Fisiología, Biología Molecular y Neurociencias, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| | - Gabriella M. Hahn
- Interdisciplinary Biomedical Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | | | - Sijung Yun
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20814
| | - Francis R. G. Amrit
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Donna B. Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA 15213
| | - Luciana F. Godoy
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| | - Esteban Salvatore
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| | - Fabiana A. Rossi
- Instituto de Investigaciones en Medicina Traslacional, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Austral, B1630 Pilar, Argentina
| | - Arjumand Ghazi
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
- Department of Cell Biology & Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - M. Cecilia Cirio
- Instituto de Fisiología, Biología Molecular y Neurociencias, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| | - Judith L. Yanowitz
- Magee-Womens Research Institute, Pittsburgh, PA 15213
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213
| | - Daniel Hochbaum
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| |
Collapse
|
10
|
daf-16/FOXO blocks adult cell fate in Caenorhabditis elegans dauer larvae via lin-41/TRIM71. PLoS Genet 2021; 17:e1009881. [PMID: 34780472 PMCID: PMC8629381 DOI: 10.1371/journal.pgen.1009881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/29/2021] [Accepted: 10/15/2021] [Indexed: 02/07/2023] Open
Abstract
Many tissue-specific stem cells maintain the ability to produce multiple cell types during long periods of non-division, or quiescence. FOXO transcription factors promote quiescence and stem cell maintenance, but the mechanisms by which FOXO proteins promote multipotency during quiescence are still emerging. The single FOXO ortholog in C. elegans, daf-16, promotes entry into a quiescent and stress-resistant larval stage called dauer in response to adverse environmental cues. During dauer, stem and progenitor cells maintain or re-establish multipotency to allow normal development to resume after dauer. We find that during dauer, daf-16/FOXO prevents epidermal stem cells (seam cells) from prematurely adopting differentiated, adult characteristics. In particular, dauer larvae that lack daf-16 misexpress collagens that are normally adult-enriched. Using col-19p::gfp as an adult cell fate marker, we find that all major daf-16 isoforms contribute to opposing col-19p::gfp expression during dauer. By contrast, daf-16(0) larvae that undergo non-dauer development do not misexpress col-19p::gfp. Adult cell fate and the timing of col-19p::gfp expression are regulated by the heterochronic gene network, including lin-41 and lin-29. lin-41 encodes an RNA-binding protein orthologous to LIN41/TRIM71 in mammals, and lin-29 encodes a conserved zinc finger transcription factor. In non-dauer development, lin-41 opposes adult cell fate by inhibiting the translation of lin-29, which directly activates col-19 transcription and promotes adult cell fate. We find that during dauer, lin-41 blocks col-19p::gfp expression, but surprisingly, lin-29 is not required in this context. Additionally, daf-16 promotes the expression of lin-41 in dauer larvae. The col-19p::gfp misexpression phenotype observed in dauer larvae with reduced daf-16 requires the downregulation of lin-41, but does not require lin-29. Taken together, this work demonstrates a novel role for daf-16/FOXO as a heterochronic gene that promotes expression of lin-41/TRIM71 to contribute to multipotent cell fate in a quiescent stem cell model. In adults and juveniles, tissue-specific stem cells divide as needed to replace cells that are lost due to injury or normal wear and tear. Many stem cells spend long periods of time in cellular quiescence, or non-division. During quiescence, stem cells remain multipotent, where they retain the ability to produce all cell types within their tissue. In this study, we define a new role for the FOXO protein DAF-16 in promoting multipotency during the quiescent C. elegans dauer larva stage. C. elegans larvae enter dauer midway through development in response to adverse environmental conditions. Epidermal stem cells are multipotent in C. elegans larvae but differentiate at adulthood, a process controlled by the “heterochronic” genes. We found that daf-16 blocks the expression of adult cell fate specifically in dauer larvae by promoting the expression of the heterochronic gene lin-41. lin-41 normally blocks adult fate by repressing the expression of another heterochronic gene, lin-29, but surprisingly, lin-29 is not needed for the expression of adult cell fate in this context. These findings may be relevant to mammals where the orthologs of daf-16 and lin-41 are important in stem cell maintenance and opposing differentiation.
Collapse
|
11
|
Basu A, Behera S, Bhardwaj S, Dey S, Ghosh-Roy A. Regulation of UNC-40/DCC and UNC-6/Netrin by DAF-16 promotes functional rewiring of the injured axon. Development 2021; 148:268990. [PMID: 34109380 DOI: 10.1242/dev.198044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 05/04/2021] [Indexed: 01/02/2023]
Abstract
The adult nervous system has a limited capacity to regenerate after accidental damage. Post-injury functional restoration requires proper targeting of the injured axon to its postsynaptic cell. Although the initial response to axonal injury has been studied in great detail, it is rather unclear what controls the re-establishment of a functional connection. Using the posterior lateral microtubule neuron in Caenorhabditis elegans, we found that after axotomy, the regrowth from the proximal stump towards the ventral side and accumulation of presynaptic machinery along the ventral nerve cord correlated to the functional recovery. We found that the loss of insulin receptor DAF-2 promoted 'ventral targeting' in a DAF-16-dependent manner. We further showed that coordinated activities of DAF-16 in neuron and muscle promoted 'ventral targeting'. In response to axotomy, expression of the Netrin receptor UNC-40 was upregulated in the injured neuron in a DAF-16-dependent manner. In contrast, the DAF-2-DAF-16 axis contributed to the age-related decline in Netrin expression in muscle. Therefore, our study revealed an important role for insulin signaling in regulating the axon guidance molecules during the functional rewiring process.
Collapse
Affiliation(s)
- Atrayee Basu
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Nainwal Mode, Gurgaon, Haryana 122051, India
| | - Sibaram Behera
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Nainwal Mode, Gurgaon, Haryana 122051, India
| | - Smriti Bhardwaj
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Nainwal Mode, Gurgaon, Haryana 122051, India
| | - Shirshendu Dey
- Fluorescence Microscopy Division, Bruker India Scientific PvT Ltd, International Trade Tower, Nehru Place, New Delhi 110019, India
| | - Anindya Ghosh-Roy
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Nainwal Mode, Gurgaon, Haryana 122051, India
| |
Collapse
|
12
|
Androwski RJ, Asad N, Wood JG, Hofer A, Locke S, Smith CM, Rose B, Schroeder NE. Mutually exclusive dendritic arbors in C. elegans neurons share a common architecture and convergent molecular cues. PLoS Genet 2020; 16:e1009029. [PMID: 32997655 PMCID: PMC7549815 DOI: 10.1371/journal.pgen.1009029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 10/12/2020] [Accepted: 08/05/2020] [Indexed: 12/31/2022] Open
Abstract
Stress-induced changes to the dendritic architecture of neurons have been demonstrated in numerous mammalian and invertebrate systems. Remodeling of dendrites varies tremendously among neuron types. During the stress-induced dauer stage of Caenorhabditis elegans, the IL2 neurons arborize to cover the anterior body wall. In contrast, the FLP neurons arborize to cover an identical receptive field during reproductive development. Using time-course imaging, we show that branching between these two neuron types is highly coordinated. Furthermore, we find that the IL2 and FLP arbors have a similar dendritic architecture and use an identical downstream effector complex to control branching; however, regulation of this complex differs between stress-induced IL2 branching and FLP branching during reproductive development. We demonstrate that the unfolded protein response (UPR) sensor IRE-1, required for localization of the complex in FLP branching, is dispensable for IL2 branching at standard cultivation temperatures. Exposure of ire-1 mutants to elevated temperatures results in defective IL2 branching, thereby demonstrating a previously unknown genotype by environment interaction within the UPR. We find that the FOXO homolog, DAF-16, is required cell-autonomously to control arborization during stress-induced arborization. Likewise, several aspects of the dauer formation pathway are necessary for the neuron to remodel, including the phosphatase PTEN/DAF-18 and Cytochrome P450/DAF-9. Finally, we find that the TOR associated protein, RAPTOR/DAF-15 regulates mutually exclusive branching of the IL2 and FLP dendrites. DAF-15 promotes IL2 branching during dauer and inhibits precocious FLP growth. Together, our results shed light on molecular processes that regulate stress-mediated remodeling of dendrites across neuron classes.
Collapse
Affiliation(s)
- Rebecca J. Androwski
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Nadeem Asad
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Janet G. Wood
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Allison Hofer
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Steven Locke
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Cassandra M. Smith
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Becky Rose
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Nathan E. Schroeder
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| |
Collapse
|
13
|
Wang C, Chen H, Li H, Zhang Y, Ren L, Chen C, Wang X, Yu J, Li Z, Liu Y. Tris(1,3-dichloro-2-propyl)phosphate Reduces the Lifespan via Activation of an Unconventional Insulin/Insulin-Like Growth Factor-1 Signaling Pathway. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:10783-10796. [PMID: 32786597 DOI: 10.1021/acs.est.0c03630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Tris(1,3-dichloro-2-propyl)phosphate (TDCPP) is an environmental contaminant that has attracted increasing concern due to its presence in environmental media and biological samples. Our previous study demonstrated that exposure to TDCPP reduced the lifespan of Caenorhabditis elegans, but the mechanisms, including the relevant signaling pathways, are unclear. The current study found that TDCPP exposure triggers an unconventional insulin/insulin-like growth factor signaling (IIS) pathway, not by disrupting the insulin-like growth factor-1 receptor DAF-2/IGF1R but by inhibiting the downstream tumor-suppressor factor DAF-18/PTEN. This inhibition reduces PI(3,4,5)P3 (PIP3) dephosphorylation, causing buildup that increases the activation of the Akt/Protein Kinase B (PKB) family of serine/threonine kinases. This activation induces DAF-16/FoxO phosphorylation and promotes the sequestration of DAF-16/FoxO in the cytoplasm, reducing the lifespan of nematodes. Our results have important diagnostic and therapeutic implications for controlling TDCPP-related diseases, especially those originating with IIS pathway components.
Collapse
Affiliation(s)
- Chen Wang
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resources and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Haibo Chen
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510655, P. R. China
| | - Hui Li
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resources and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
| | - Yunchao Zhang
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resources and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Luyao Ren
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resources and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Chao Chen
- State Key Laboratory of Bioreactor Engineering, Biomedical Nanotechnology Center, Shanghai Collaborative Innovation Center for Biomanufacturing, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Xiaoli Wang
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resources and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Jun Yu
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resources and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Zongrui Li
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510655, P. R. China
| | - Yongdi Liu
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resources and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| |
Collapse
|
14
|
Mei ZG, Huang YG, Feng ZT, Luo YN, Yang SB, Du LP, Jiang K, Liu XL, Fu XY, Deng YH, Zhou HJ. Electroacupuncture ameliorates cerebral ischemia/reperfusion injury by suppressing autophagy via the SIRT1-FOXO1 signaling pathway. Aging (Albany NY) 2020; 12:13187-13205. [PMID: 32620714 PMCID: PMC7377856 DOI: 10.18632/aging.103420] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
Cerebral ischemia/reperfusion (CIR) injury occurs when blood flow is restored in the brain, causing secondary damage to the ischemic tissues. Previous studies have shown that electroacupuncture (EA) treatment contributes to brain protection against CIR injury through modulating autophagy. Studies indicated that SIRT1-FOXO1 plays a crucial role in regulating autophagy. Here we investigated the mechanisms underlying the neuroprotective effect of EA and its role in modulating autophagy via the SIRT1-FOXO1 signaling pathway in rats with CIR injury. EA pretreatment at "Baihui", "Quchi" and "Zusanli" acupoints (2/15Hz, 1mA, 30 min/day) was performed for 5 days before the rats were subjected to middle cerebral artery occlusion, and the results indicated that EA pretreatment substantially reduced the Longa score and infarct volume, increased the dendritic spine density and lessened autophagosomes in the peri-ischemic cortex of rats. Additionally, EA pretreatment also reduced the ratio of LC3-II/LC3-I, the levels of Ac-FOXO1 and Atg7, and the interaction of Ac-FOXO1 and Atg7, but increased the levels of p62, SIRT1, and FOXO1. The above effects were abrogated by the SIRT1 inhibitor EX527. Thus, we presume that EA pretreatment elicits a neuroprotective effect against CIR injury, potentially by suppressing autophagy via activating the SIRT1-FOXO1 signaling pathway.
Collapse
Affiliation(s)
- Zhi-Gang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, China
| | - Ya-Guang Huang
- Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Zhi-Tao Feng
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, China
| | - Ya-Nan Luo
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, China
| | - Song-Bai Yang
- Yichang Hospital of Traditional Chinese Medicine, Clinical Medical College of Traditional Chinese Medicine, China Three Gorges University, Yichang, Hubei, China
| | - Li-Peng Du
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, China
| | - Kang Jiang
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, China
| | - Xiao-Lu Liu
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, China
| | - Xian-Yun Fu
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, China
| | - Yi-Hui Deng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hua-Jun Zhou
- The Institute of Neurology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
15
|
Christensen EL, Beasley A, Radchuk J, Mielko ZE, Preston E, Stuckett S, Murray JI, Hudson ML. ngn-1/neurogenin Activates Transcription of Multiple Terminal Selector Transcription Factors in the Caenorhabditis elegans Nervous System. G3 (BETHESDA, MD.) 2020; 10:1949-1962. [PMID: 32273286 PMCID: PMC7263688 DOI: 10.1534/g3.120.401126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/30/2020] [Indexed: 11/18/2022]
Abstract
Proper nervous system development is required for an organism's survival and function. Defects in neurogenesis have been linked to neurodevelopmental disorders such as schizophrenia and autism. Understanding the gene regulatory networks that orchestrate neural development, specifically cascades of proneural transcription factors, can better elucidate which genes are most important during early neurogenesis. Neurogenins are a family of deeply conserved factors shown to be both necessary and sufficient for the development of neural subtypes. However, the immediate downstream targets of neurogenin are not well characterized. The objective of this study was to further elucidate the role of ngn-1/neurogenin in nervous system development and to identify its downstream transcriptional targets, using the nematode Caenorhabditis elegans as a model for this work. We found that ngn-1 is required for axon outgrowth, nerve ring architecture, and neuronal cell fate specification. We also showed that ngn-1 may have roles in neuroblast migration and epithelial integrity during embryonic development. Using RNA sequencing and comparative transcriptome analysis, we identified eight transcription factors (hlh-34/NPAS1, unc-42/PROP1, ceh-17/PHOX2A, lim-4/LHX6, fax-1/NR2E3, lin-11/LHX1, tlp-1/ZNF503, and nhr-23/RORB) whose transcription is activated, either directly or indirectly, by ngn-1 Our results show that ngn-1 has a role in transcribing known terminal regulators that establish and maintain cell fate of differentiated neural subtypes and confirms that ngn-1 functions as a proneural transcription factor in C. elegans neurogenesis.
Collapse
Affiliation(s)
- Elyse L Christensen
- Department of Molecular and Cellular Biology, Kennesaw State University, GA 30144
| | - Alexandra Beasley
- Department of Molecular and Cellular Biology, Kennesaw State University, GA 30144
| | - Jessica Radchuk
- Department of Molecular and Cellular Biology, Kennesaw State University, GA 30144
| | - Zachery E Mielko
- Department of Molecular and Cellular Biology, Kennesaw State University, GA 30144
| | - Elicia Preston
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Sidney Stuckett
- Department of Molecular and Cellular Biology, Kennesaw State University, GA 30144
| | - John I Murray
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Martin L Hudson
- Department of Molecular and Cellular Biology, Kennesaw State University, GA 30144
| |
Collapse
|
16
|
Novakovic S, Molesworth LW, Gourley TE, Boag PR, Davis GM. Zinc transporters maintain longevity by influencing insulin/IGF-1 activity in Caenorhabditis elegans. FEBS Lett 2020; 594:1424-1432. [PMID: 31883120 DOI: 10.1002/1873-3468.13725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/05/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
Adequate dietary intake of essential metals such as zinc is important for maintaining homeostasis. Abnormal zinc intake in Caenorhabditis elegans has been shown to increase or decrease normal lifespan by influencing the insulin/IGF-1 pathway. Distribution of zinc is achieved by a family of highly conserved zinc transport proteins (ZIPT in C. elegans). This study investigated the role of the zipt family of genes and showed that depletion of individual zipt genes results in a decreased lifespan. Moreover, zipt-16 and zipt-17 mutants synthetically interact with the insulin/IGF cofactors daf-16 and skn-1, and cause abnormal localisation of DAF-16. This study suggests that the zipt family of genes are required for maintaining normal lifespan through influencing the insulin/IGF-1 pathway.
Collapse
Affiliation(s)
- Stevan Novakovic
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Luke W Molesworth
- School of Health and Life Sciences, Federation University, Churchill, VIC, Australia
| | - Taylin E Gourley
- School of Health and Life Sciences, Federation University, Churchill, VIC, Australia
| | - Peter R Boag
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Gregory M Davis
- School of Health and Life Sciences, Federation University, Churchill, VIC, Australia
| |
Collapse
|
17
|
DAF-16/FoxO in Caenorhabditis elegans and Its Role in Metabolic Remodeling. Cells 2020; 9:cells9010109. [PMID: 31906434 PMCID: PMC7017163 DOI: 10.3390/cells9010109] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 12/31/2022] Open
Abstract
DAF-16, the only forkhead box transcription factors class O (FoxO) homolog in Caenorhabditis elegans, integrates signals from upstream pathways to elicit transcriptional changes in many genes involved in aging, development, stress, metabolism, and immunity. The major regulator of DAF-16 activity is the insulin/insulin-like growth factor 1 (IGF-1) signaling (IIS) pathway, reduction of which leads to lifespan extension in worms, flies, mice, and humans. In C. elegans daf-2 mutants, reduced IIS leads to a heterochronic activation of a dauer survival program during adulthood. This program includes elevated antioxidant defense and a metabolic shift toward accumulation of carbohydrates (i.e., trehalose and glycogen) and triglycerides, and activation of the glyoxylate shunt, which could allow fat-to-carbohydrate conversion. The longevity of daf-2 mutants seems to be partially supported by endogenous trehalose, a nonreducing disaccharide that mammals cannot synthesize, which points toward considerable differences in downstream mechanisms by which IIS regulates aging in distinct groups.
Collapse
|
18
|
Nagashima T, Iino Y, Tomioka M. DAF-16/FOXO promotes taste avoidance learning independently of axonal insulin-like signaling. PLoS Genet 2019; 15:e1008297. [PMID: 31323047 PMCID: PMC6668909 DOI: 10.1371/journal.pgen.1008297] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 07/31/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022] Open
Abstract
The avoidance of starvation is critical for the survival of most organisms, thus animals change behavior based on past nutritional conditions. Insulin signaling is important for nutritional state-dependent behavioral plasticity, yet the underlying regulatory mechanism at the cellular level remains unclear. Previous studies showed that insulin-like signaling is required for taste avoidance learning, in which the nematode Caenorhabditis elegans avoids salt concentrations encountered under starvation conditions. DAF-2c, a splice isoform of the DAF-2 insulin receptor, functions in the axon of the ASER sensory neuron, which senses changes in salt concentrations. In addition, mutants of a major downstream factor of DAF-2, the forkhead transcription factor O (FOXO) homolog DAF-16, show defects in taste avoidance learning. Interestingly, the defect of the daf-2 mutant is not suppressed by daf-16 mutations in the learning, unlike those in other phenomena, such as longevity and development. Here we show that multiple DAF-16 isoforms function in ASER. By epistasis analysis using a DAF-2c isoform-specific mutant and an activated form of DAF-16, we found that DAF-16 acts in the nucleus in parallel with the DAF-2c-dependent pathway in the axon, indicating that insulin-like signaling acts both in the cell body and axon of a single neuron, ASER. Starvation conditioning induces nuclear translocation of DAF-16 in ASER and degradation of DAF-16 before starvation conditioning causes defects in taste avoidance learning. Forced nuclear localization of DAF-16 in ASER biased chemotaxis towards lower salt concentrtions and this effect required the Gq/PKC pathway and neuropeptide processing enzymes. These data imply that DAF-16/FOXO transmits starvation signals and modulates neuropeptide transmission in the learning. Animals change behavior based on remembered experiences of hunger and appetite. Signaling by insulin and insulin-like peptides in the nervous system plays key roles in behavioral responses to hunger and satiety. In C. elegans, insulin-like signaling in the gustatory sensory neuron ASER regulates learned avoidance of salt concentrations experienced during fasting, which we call taste avoidance learning. DAF-2c, an isoform of the insulin receptor homolog, is localized to the axon of ASER and regulates taste avoidance learning. Here, we show that DAF-16, the forkhead transcription factor O (FOXO) homolog, translocates into the nucleus of ASER during fasting and promotes taste avoidance learning. DAF-16 is negatively regulated by insulin-like signaling independently of axonal DAF-2c signaling. This dual function of insulin-like signaling in the cell body and the axon ensures dynamic changes in behavioral responses after experience of hunger. By genetic analyses using constitutively nuclear-translocated DAF-16, we show that DAF-16 in ASER regulates taste avoidance learning via modulating neuropeptide signaling in the nervous system, which is reminiscent of the function of FOXO in the hypothalamus in the regulation of food-seeking behavior in mammals.
Collapse
Affiliation(s)
- Takashi Nagashima
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Yuichi Iino
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Masahiro Tomioka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
19
|
daf-16/FOXO isoform b in AIY neurons is involved in low preference for Bifidobacterium infantis in Caenorhabditis elegans. Neurosci Res 2019; 150:8-16. [PMID: 30731110 DOI: 10.1016/j.neures.2019.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 02/01/2023]
Abstract
The neural and molecular mechanisms underlying food preference have been poorly understood. We previously showed that Bifidobacterium infantis (B. infantis), a well-known probiotic bacterium, extends the lifespan of Caenorhabditis elegans (C. elegans) compared with a standard food, Escherichia coli (E. coli) OP50. In this study, we characterized C. elegans behavior against B. infantis and examined the neural and molecular mechanisms governing that behavior. The majority of the wild-type animals were outside of the B. infantis lawn 10 min after transfer. Although worms did not prefer B. infantis compared to E. coli OP50, they preferred the B. infantis lawn over a lawn containing M9 buffer alone, in which there was no food. Mutant analyses suggested that leaving the B. infantis lawn required daf-16/FOXO. Isoform-specific mutant phenotypes suggested that daf-16 isoform b seemed to be associated with leaving. Genetic rescue experiments demonstrated that the function of daf-16b in AIY interneurons was involved in leaving the B. infantis lawn. The daf-18/PTEN mutants were also defective in leaving. In conclusion, C. elegans showed a low preference for B. infantis, and daf-16b in AIY interneurons and daf-18 had roles in leaving B. infantis.
Collapse
|
20
|
Abstract
Recent reports emphasized the role of FOXO family of transcription factors in nervous system homeostasis. Most studies employed primary neuronal cultures, established animal models for neuropathology, or invertebrate models. Demonstration of the normal and pathophysiological function of mammalian FOXO under complex in vivo conditions requires genetic study. Therefore, the conditional knockout mouse is an invaluable platform. Here, we describe the methods of establishing and analyzing nervous system-specific ablation of FOXO isoforms in mice. This chapter offers a detailed method to validate the deletion of Foxo genes in vivo and to study its role in the nervous system. Investigation of FOXO function by using the mouse system may advance our understanding of nervous system aging as well as neurodegenerative diseases.
Collapse
Affiliation(s)
- Hwanhee Oh
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jihye Paik
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
21
|
McLaughlin CN, Broihier HT. Keeping Neurons Young and Foxy: FoxOs Promote Neuronal Plasticity. Trends Genet 2018; 34:65-78. [PMID: 29102406 DOI: 10.1016/j.tig.2017.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 12/27/2022]
Abstract
Any adult who has tried to take up the piano or learn a new language is faced with the sobering realization that acquiring such skills is more challenging as an adult than as a child. Neuronal plasticity, or the malleability of brain circuits, declines with age. Young neurons tend to be more adaptable and can alter the size and strength of their connections more readily than can old neurons. Myriad circuit- and synapse-level mechanisms that shape plasticity have been identified. Yet, molecular mechanisms setting the overall competence of young neurons for distinct forms of plasticity remain largely obscure. Recent studies indicate evolutionarily conserved roles for FoxO proteins in establishing the capacity for cell-fate, morphological, and synaptic plasticity in neurons.
Collapse
Affiliation(s)
- Colleen N McLaughlin
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Heather T Broihier
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
22
|
Somogyvári M, Gecse E, Sőti C. DAF-21/Hsp90 is required for C. elegans longevity by ensuring DAF-16/FOXO isoform A function. Sci Rep 2018; 8:12048. [PMID: 30104664 PMCID: PMC6089956 DOI: 10.1038/s41598-018-30592-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 08/02/2018] [Indexed: 01/25/2023] Open
Abstract
The FOXO transcription factor family is a conserved regulator of longevity and the downstream target of insulin/insulin-like signaling. In Caenorhabditis elegans, the FOXO ortholog DAF-16A and D/F isoforms extend lifespan in daf-2 insulin-like receptor mutants. Here we identify the DAF-21/Hsp90 chaperone as a longevity regulator. We find that reducing DAF-21 capacity by daf-21(RNAi) initiated either at the beginning or at the end of larval development shortens wild-type lifespan. daf-21 knockdown employed from the beginning of larval development also decreases longevity of daf-2 mutant and daf-2 silenced nematodes. daf-16 loss-of-function mitigates the lifespan shortening effect of daf-21 silencing. We demonstrate that DAF-21 specifically promotes daf-2 and heat-shock induced nuclear translocation of DAF-16A as well as the induction of DAF-16A-specific mRNAs, without affecting DAF-16D/F localization and transcriptional function. DAF-21 is dispensable for the stability and nuclear import of DAF-16A, excluding a chaperone-client interaction and suggesting that DAF-21 regulates DAF-16A activation upstream of its cellular traffic. Finally, we show a selective requirement for DAF-21 to extend lifespan of DAF-16A, but not DAF-16D/F, transgenic daf-2 mutant strains. Our findings indicate a spatiotemporal determination of multiple DAF-21 roles in fertility, development and longevity and reveal an isoform-specific regulation of DAF-16 activity.
Collapse
Affiliation(s)
- Milán Somogyvári
- Department of Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Eszter Gecse
- Department of Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Csaba Sőti
- Department of Medical Chemistry, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
23
|
Abstract
In Caenorhabditis elegans, there is a single FOXO transcription factor homolog, encoded by the gene, daf-16. As a central regulator for multiple signaling pathways, DAF-16 integrates these signals which results in modulation of several biological processes including longevity, development, fat storage, stress resistance, innate immunity, and reproduction. Using C. elegans allows for studies of FOXO in the context of the whole animal. Therefore, manipulating levels or the activity of daf-16 results in phenotypic changes. Genetic and molecular analysis revealed that similar to other systems, DAF-16 is the downstream target of the conserved insulin/IGF-1 signaling (IIS) pathway; a PI 3-kinase/AKT signaling cascade that ultimately controls the regulation of DAF-16 nuclear localization. In this chapter, I will focus on understanding how a single gene daf-16 can incorporate signals from multiple upstream pathways and in turn modulate different phenotypes as well as the study of FOXO in the context of a whole organism.
Collapse
Affiliation(s)
- Heidi A Tissenbaum
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, United States.
| |
Collapse
|
24
|
Hwang I, Oh H, Santo E, Kim DY, Chen JW, Bronson RT, Locasale JW, Na Y, Lee J, Reed S, Toth M, Yu WH, Muller FL, Paik J. FOXO protects against age-progressive axonal degeneration. Aging Cell 2018; 17. [PMID: 29178390 PMCID: PMC5771393 DOI: 10.1111/acel.12701] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2017] [Indexed: 12/16/2022] Open
Abstract
Neurodegeneration resulting in cognitive and motor impairment is an inevitable consequence of aging. Little is known about the genetic regulation of this process despite its overriding importance in normal aging. Here, we identify the Forkhead Box O (FOXO) transcription factor 1, 3, and 4 isoforms as a guardian of neuronal integrity by inhibiting age-progressive axonal degeneration in mammals. FOXO expression progressively increased in aging human and mouse brains. The nervous system-specific deletion of Foxo transcription factors in mice accelerates aging-related axonal tract degeneration, which is followed by motor dysfunction. This accelerated neurodegeneration is accompanied by levels of white matter astrogliosis and microgliosis in middle-aged Foxo knockout mice that are typically only observed in very old wild-type mice and other aged mammals, including humans. Mechanistically, axonal degeneration in nerve-specific Foxo knockout mice is associated with elevated mTORC1 activity and accompanying proteotoxic stress due to decreased Sestrin3 expression. Inhibition of mTORC1 by rapamycin treatment mimics FOXO action and prevented axonal degeneration in Foxo knockout mice with accelerated nervous system aging. Defining this central role for FOXO in neuroprotection during mammalian aging offers an invaluable window into the aging process itself.
Collapse
Affiliation(s)
- Inah Hwang
- Department of Pathology and Laboratory Medicine; Weill Cornell Medicine; New York NY USA
| | - Hwanhee Oh
- Department of Pathology and Laboratory Medicine; Weill Cornell Medicine; New York NY USA
| | - Evan Santo
- Department of Pathology and Laboratory Medicine; Weill Cornell Medicine; New York NY USA
| | - Do-Yeon Kim
- Department of Pharmacology; School of Dentistry; Kyungpook National University; Daegu Korea
| | - John W. Chen
- Center for Systems Biology and the Division of Neuroradiology; Department of Radiology; Massachusetts General Hospital; Harvard Medical School; Boston MA USA
| | - Roderick T. Bronson
- Department of Microbiology and Immunobiology; Harvard Medical School; Boston MA USA
| | - Jason W. Locasale
- Department of Pharmacology and Cancer Biology; Duke University School of Medicine; Durham NC USA
| | - Yoonmi Na
- Department of Pathology and Laboratory Medicine; Weill Cornell Medicine; New York NY USA
| | - Jaclyn Lee
- Department of Medical Oncology; Dana Farber Cancer Institute; Boston MA USA
| | - Stewart Reed
- Department of Medical Oncology; Dana Farber Cancer Institute; Boston MA USA
| | - Miklos Toth
- Department of Pharmacology; Weill Cornell Medicine; New York NY USA
| | - Wai H. Yu
- Department of Pathology and Cell Biology; Columbia University; New York NY USA
| | - Florian L. Muller
- Cancer Systems Imaging; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Jihye Paik
- Department of Pathology and Laboratory Medicine; Weill Cornell Medicine; New York NY USA
| |
Collapse
|
25
|
Ledda F, Paratcha G. Mechanisms regulating dendritic arbor patterning. Cell Mol Life Sci 2017; 74:4511-4537. [PMID: 28735442 PMCID: PMC11107629 DOI: 10.1007/s00018-017-2588-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 06/14/2017] [Accepted: 07/06/2017] [Indexed: 12/17/2022]
Abstract
The nervous system is populated by diverse types of neurons, each of which has dendritic trees with strikingly different morphologies. These neuron-specific morphologies determine how dendritic trees integrate thousands of synaptic inputs to generate different firing properties. To ensure proper neuronal function and connectivity, it is necessary that dendrite patterns are precisely controlled and coordinated with synaptic activity. Here, we summarize the molecular and cellular mechanisms that regulate the formation of cell type-specific dendrite patterns during development. We focus on different aspects of vertebrate dendrite patterning that are particularly important in determining the neuronal function; such as the shape, branching, orientation and size of the arbors as well as the development of dendritic spine protrusions that receive excitatory inputs and compartmentalize postsynaptic responses. Additionally, we briefly comment on the implications of aberrant dendritic morphology for nervous system disease.
Collapse
Affiliation(s)
- Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Paraguay 2155, 3rd Floor, CABA, 1121, Buenos Aires, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Paraguay 2155, 3rd Floor, CABA, 1121, Buenos Aires, Argentina.
| |
Collapse
|
26
|
Abstract
The FOXO family of transcription factors plays a conserved role in longevity and tissue homeostasis across species. In the mammalian nervous system, emerging evidence has implicated FOXOs in cognitive performance, stem cell maintenance, regeneration, and protection against stress. Much of what we know about neuronal functions of FOXO emerged from recent studies in C. elegans. Similar to mammalian FOXO, the worm FOXO ortholog, called DAF-16, regulates learning and memory, regeneration, and stress resistance in neurons. Here, we discuss the current state of our knowledge of FOXO’s functions in neurons in mammals and invertebrates, and highlight areas where our understanding is limited. Defining the function of FOXO factors in the healthy, aged, and diseased brain may have important implications for improving healthspan and treating neurodegenerative disease.
Collapse
Affiliation(s)
- Sun Y Kim
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| |
Collapse
|
27
|
Chisholm AD, Hutter H, Jin Y, Wadsworth WG. The Genetics of Axon Guidance and Axon Regeneration in Caenorhabditis elegans. Genetics 2016; 204:849-882. [PMID: 28114100 PMCID: PMC5105865 DOI: 10.1534/genetics.115.186262] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/06/2016] [Indexed: 11/18/2022] Open
Abstract
The correct wiring of neuronal circuits depends on outgrowth and guidance of neuronal processes during development. In the past two decades, great progress has been made in understanding the molecular basis of axon outgrowth and guidance. Genetic analysis in Caenorhabditis elegans has played a key role in elucidating conserved pathways regulating axon guidance, including Netrin signaling, the slit Slit/Robo pathway, Wnt signaling, and others. Axon guidance factors were first identified by screens for mutations affecting animal behavior, and by direct visual screens for axon guidance defects. Genetic analysis of these pathways has revealed the complex and combinatorial nature of guidance cues, and has delineated how cues guide growth cones via receptor activity and cytoskeletal rearrangement. Several axon guidance pathways also affect directed migrations of non-neuronal cells in C. elegans, with implications for normal and pathological cell migrations in situations such as tumor metastasis. The small number of neurons and highly stereotyped axonal architecture of the C. elegans nervous system allow analysis of axon guidance at the level of single identified axons, and permit in vivo tests of prevailing models of axon guidance. C. elegans axons also have a robust capacity to undergo regenerative regrowth after precise laser injury (axotomy). Although such axon regrowth shares some similarities with developmental axon outgrowth, screens for regrowth mutants have revealed regeneration-specific pathways and factors that were not identified in developmental screens. Several areas remain poorly understood, including how major axon tracts are formed in the embryo, and the function of axon regeneration in the natural environment.
Collapse
Affiliation(s)
| | - Harald Hutter
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, V5A 1S6, Canada
| | - Yishi Jin
- Section of Neurobiology, Division of Biological Sciences, and
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093
- Department of Pathology and Laboratory Medicine, Howard Hughes Medical Institute, Chevy Chase, Maryland, and
| | - William G Wadsworth
- Department of Pathology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| |
Collapse
|
28
|
Colón-Ramos DA. The need to connect: on the cell biology of synapses, behaviors, and networks in science. Mol Biol Cell 2016; 27:3197-3201. [PMID: 27799494 PMCID: PMC5170851 DOI: 10.1091/mbc.e16-07-0507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
My laboratory is interested in the cell biology of the synapse. Synapses, which are points of cellular communication between neurons, were first described by Santiago Ramón y Cajal as "protoplasmic kisses that appear to constitute the final ecstasy of an epic love story." Who would not want to work on that?! My lab examines the biological mechanisms neurons use to find and connect to each other. How are synapses formed during development, maintained during growth, and modified during learning? In this essay, I reflect about my scientific journey to the synapse, the cell biological one, but also a metaphorical synapse-my role as a point of contact between the production of knowledge and its dissemination. In particular, I discuss how the architecture of scientific networks propels knowledge production but can also exclude certain groups in science.
Collapse
Affiliation(s)
- Daniel A Colón-Ramos
- Department of Cell Biology and Neuroscience, Yale School of Medicine, New Haven, CT 06510; Instituto de Neurobiología, Recinto de Ciencias Médicas, Universidad de Puerto Rico, San Juan, PR 00901
| |
Collapse
|
29
|
Sears JC, Broihier HT. FoxO regulates microtubule dynamics and polarity to promote dendrite branching in Drosophila sensory neurons. Dev Biol 2016; 418:40-54. [PMID: 27546375 DOI: 10.1016/j.ydbio.2016.08.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 08/12/2016] [Accepted: 08/16/2016] [Indexed: 01/15/2023]
Abstract
The size and shape of dendrite arbors are defining features of neurons and critical determinants of neuronal function. The molecular mechanisms establishing arborization patterns during development are not well understood, though properly regulated microtubule (MT) dynamics and polarity are essential. We previously found that FoxO regulates axonal MTs, raising the question of whether it also regulates dendritic MTs and morphology. Here we demonstrate that FoxO promotes dendrite branching in all classes of Drosophila dendritic arborization (da) neurons. FoxO is required both for initiating growth of new branches and for maintaining existing branches. To elucidate FoxO function, we characterized MT organization in both foxO null and overexpressing neurons. We find that FoxO directs MT organization and dynamics in dendrites. Moreover, it is both necessary and sufficient for anterograde MT polymerization, which is known to promote dendrite branching. Lastly, FoxO promotes proper larval nociception, indicating a functional consequence of impaired da neuron morphology in foxO mutants. Together, our results indicate that FoxO regulates dendrite structure and function and suggest that FoxO-mediated pathways control MT dynamics and polarity.
Collapse
Affiliation(s)
- James C Sears
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Heather T Broihier
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
30
|
O'Connor-Giles K. Toll-tally tubular: A newly identified Toll-like receptor-FoxO pathway regulates dynamics of the neuronal microtubule network. J Cell Biol 2016; 214:371-3. [PMID: 27528655 PMCID: PMC4987299 DOI: 10.1083/jcb.201607118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 11/22/2022] Open
Abstract
Recent studies reveal a conserved role for FoxO transcription factors in establishing neuronal structure and circuit function. In this issue, McLaughlin et al. (2016. J. Cell Biol. http://dx.doi.org/10.1083/jcb.201601014) identify a novel Toll-like receptor-FoxO pathway that represses the mitotic kinesin Pavarotti/MKLP1 to promote dynamic microtubules required for axonal transport and activity-dependent remodeling of presynaptic terminals.
Collapse
|
31
|
McLaughlin CN, Nechipurenko IV, Liu N, Broihier HT. A Toll receptor-FoxO pathway represses Pavarotti/MKLP1 to promote microtubule dynamics in motoneurons. J Cell Biol 2016; 214:459-74. [PMID: 27502486 PMCID: PMC4987293 DOI: 10.1083/jcb.201601014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/07/2016] [Indexed: 02/06/2023] Open
Abstract
McLaughlin et al. uncover a motoneuronal Toll-6–directed pathway that functions via dSARM and FoxO to attenuate microtubule stability through repression of Pavarotti/MKLP1 transcription. Genetic and pharmacological strategies reveal a novel requirement for dynamic synaptic microtubules in structural plasticity, which are established by Toll-6–FoxO signaling. FoxO proteins are evolutionarily conserved regulators of neuronal structure and function, yet the neuron-specific pathways within which they act are poorly understood. To elucidate neuronal FoxO function in Drosophila melanogaster, we first screened for FoxO’s upstream regulators and downstream effectors. On the upstream side, we present genetic and molecular pathway analyses indicating that the Toll-6 receptor, the Toll/interleukin-1 receptor domain adaptor dSARM, and FoxO function in a linear pathway. On the downstream side, we find that Toll-6–FoxO signaling represses the mitotic kinesin Pavarotti/MKLP1 (Pav-KLP), which itself attenuates microtubule (MT) dynamics. We next probed in vivo functions for this novel pathway and found that it is essential for axon transport and structural plasticity in motoneurons. We demonstrate that elevated expression of Pav-KLP underlies transport and plasticity phenotypes in pathway mutants, indicating that Toll-6–FoxO signaling promotes MT dynamics by limiting Pav-KLP expression. In addition to uncovering a novel molecular pathway, our work reveals an unexpected function for dynamic MTs in enabling rapid activity-dependent structural plasticity.
Collapse
Affiliation(s)
- Colleen N McLaughlin
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| | - Inna V Nechipurenko
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| | - Nan Liu
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| | - Heather T Broihier
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
32
|
The Caenorhabditis elegans Ephrin EFN-4 Functions Non-cell Autonomously with Heparan Sulfate Proteoglycans to Promote Axon Outgrowth and Branching. Genetics 2015; 202:639-60. [PMID: 26645816 DOI: 10.1534/genetics.115.185298] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/02/2015] [Indexed: 01/21/2023] Open
Abstract
The Eph receptors and their cognate ephrin ligands play key roles in many aspects of nervous system development. These interactions typically occur within an individual tissue type, serving either to guide axons to their terminal targets or to define boundaries between the rhombomeres of the hindbrain. We have identified a novel role for the Caenorhabditis elegans ephrin EFN-4 in promoting primary neurite outgrowth in AIY interneurons and D-class motor neurons. Rescue experiments reveal that EFN-4 functions non-cell autonomously in the epidermis to promote primary neurite outgrowth. We also find that EFN-4 plays a role in promoting ectopic axon branching in a C. elegans model of X-linked Kallmann syndrome. In this context, EFN-4 functions non-cell autonomously in the body-wall muscle and in parallel with HS modification genes and HSPG core proteins. This is the first report of an epidermal ephrin providing a developmental cue to the nervous system.
Collapse
|
33
|
Hesp K, Smant G, Kammenga JE. Caenorhabditis elegans DAF-16/FOXO transcription factor and its mammalian homologs associate with age-related disease. Exp Gerontol 2015; 72:1-7. [DOI: 10.1016/j.exger.2015.09.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/02/2015] [Accepted: 09/06/2015] [Indexed: 11/29/2022]
|
34
|
DNA methylation changes in the placenta are associated with fetal manganese exposure. Reprod Toxicol 2015; 57:43-9. [PMID: 25982381 DOI: 10.1016/j.reprotox.2015.05.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 04/13/2015] [Accepted: 05/07/2015] [Indexed: 12/22/2022]
Abstract
Adequate micronutrient intake, including manganese (Mn), is important for fetal development. Both Mn deficiencies and excess exposures are associated with later-life disease, and Mn accumulates in the placenta. Placental functional alterations may alter fetal programming and lifelong health, and we hypothesized that prenatal exposures to Mn may alter placental function through epigenetic mechanisms. Using Illumina's HumanMethylation450 BeadArray, DNA methylation of >485,000 CpG loci genome-wide was interrogated in 61 placental samples and Mn associations assessed genome-wide via omnibus test (p=0.045). 713 loci were associated with Mn exposure (p<0.0001). Five significantly differentially-methylated (p<1.3×10(-7)) loci reside in neurodevelopmental, fetal growth and cancer-related genes. cg22284422, within the uncharacterized LOC284276 gene, was associated with birth weight; for every 10% increase in methylation, lower birth weights were observed, with an average decrease of 293.44g. Our observations suggest a link between prenatal micronutrient levels, placental epigenetic status and birth weight, although these preliminary results require validation.
Collapse
|
35
|
Liu J, Chin-Sang ID. C. elegans as a model to study PTEN's regulation and function. Methods 2014; 77-78:180-90. [PMID: 25514044 DOI: 10.1016/j.ymeth.2014.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 12/04/2014] [Accepted: 12/05/2014] [Indexed: 02/06/2023] Open
Abstract
PTEN (phosphatase and tensin homolog deleted on chromosome 10) has important roles in tumor suppression, metabolism, and development, yet its regulators, effectors, and functions are not fully understood. DAF-18 is the PTEN ortholog in Caenorhabditis elegans. DAF-18's role is highly conserved to human PTEN, and can be functionally replaced by human PTEN. Thus C. elegans provides a valuable model to study PTEN. This review assesses current and emerging methods to study DAF-18's regulators and functions in C. elegans. We propose genetic modify screens to identify genes that interact with daf-18/PTEN. These genes are potential targets for anticancer drug therapies. We also provide a review on the roles DAF-18/PTEN has during C. elegans development and how studying these physiological roles can provide mechanistic insight on DAF-18/PTEN function.
Collapse
Affiliation(s)
- Jun Liu
- Department of Biology, Queen's University, 116 Barrie St., Kingston, ON K7L 3N6, Canada.
| | - Ian D Chin-Sang
- Department of Biology, Queen's University, 116 Barrie St., Kingston, ON K7L 3N6, Canada.
| |
Collapse
|
36
|
Gkikas I, Petratou D, Tavernarakis N. Longevity pathways and memory aging. Front Genet 2014; 5:155. [PMID: 24926313 PMCID: PMC4044971 DOI: 10.3389/fgene.2014.00155] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 05/10/2014] [Indexed: 12/28/2022] Open
Abstract
The aging process has been associated with numerous pathologies at the cellular, tissue, and organ level. Decline or loss of brain functions, including learning and memory, is one of the most devastating and feared aspects of aging. Learning and memory are fundamental processes by which animals adjust to environmental changes, evaluate various sensory signals based on context and experience, and make decisions to generate adaptive behaviors. Age-related memory impairment is an important phenotype of brain aging. Understanding the molecular mechanisms underlying age-related memory impairment is crucial for the development of therapeutic strategies that may eventually lead to the development of drugs to combat memory loss. Studies in invertebrate animal models have taught us much about the physiology of aging and its effects on learning and memory. In this review we survey recent progress relevant to conserved molecular pathways implicated in both aging and memory formation and consolidation.
Collapse
Affiliation(s)
- Ilias Gkikas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion Crete, Greece
| | - Dionysia Petratou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion Crete, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion Crete, Greece ; Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion Crete, Greece
| |
Collapse
|
37
|
Veleva-Rotse BO, Barnes AP. Brain patterning perturbations following PTEN loss. Front Mol Neurosci 2014; 7:35. [PMID: 24860420 PMCID: PMC4030135 DOI: 10.3389/fnmol.2014.00035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 04/15/2014] [Indexed: 12/23/2022] Open
Abstract
This review will consider the impact of compromised PTEN signaling in brain patterning. We approach understanding the contribution of PTEN to nervous system development by surveying the findings from the numerous genetic loss-of-function models that have been generated as well as other forms of PTEN inactivation. By exploring the developmental programs influenced by this central transduction molecule, we can begin to understand the molecular mechanisms that shape the developing brain. A wealth of data indicates that PTEN plays critical roles in a variety of stages during brain development. Many of them are considered here including: stem cell proliferation, fate determination, polarity, migration, process outgrowth, myelination and somatic hypertrophy. In many of these contexts, it is clear that PTEN phosphatase activity contributes to the observed effects of genetic deletion or depletion, however recent studies have also ascribed non-catalytic functions to PTEN in regulating cell function. We also explore the potential impact this alternative pool of PTEN may have on the developing brain. Together, these elements begin to form a clearer picture of how PTEN contributes to the emergence of brain structure and binds form and function in the nervous system.
Collapse
Affiliation(s)
- Biliana O Veleva-Rotse
- Neuroscience Graduate Program, Oregon Health and Science University Portland, OR, USA ; Department of Pediatrics, Oregon Health and Science University Portland, OR, USA
| | - Anthony P Barnes
- Neuroscience Graduate Program, Oregon Health and Science University Portland, OR, USA ; Department of Pediatrics, Oregon Health and Science University Portland, OR, USA ; Department of Cell and Developmental Biology, Oregon Health and Science University Portland, OR, USA
| |
Collapse
|
38
|
Namba T, Kibe Y, Funahashi Y, Nakamuta S, Takano T, Ueno T, Shimada A, Kozawa S, Okamoto M, Shimoda Y, Oda K, Wada Y, Masuda T, Sakakibara A, Igarashi M, Miyata T, Faivre-Sarrailh C, Takeuchi K, Kaibuchi K. Pioneering axons regulate neuronal polarization in the developing cerebral cortex. Neuron 2014; 81:814-29. [PMID: 24559674 DOI: 10.1016/j.neuron.2013.12.015] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2013] [Indexed: 01/06/2023]
Abstract
The polarization of neurons, which mainly includes the differentiation of axons and dendrites, is regulated by cell-autonomous and non-cell-autonomous factors. In the developing central nervous system, neuronal development occurs in a heterogeneous environment that also comprises extracellular matrices, radial glial cells, and neurons. Although many cell-autonomous factors that affect neuronal polarization have been identified, the microenvironmental cues involved in neuronal polarization remain largely unknown. Here, we show that neuronal polarization occurs in a microenvironment in the lower intermediate zone, where the cell adhesion molecule transient axonal glycoprotein-1 (TAG-1) is expressed in cortical efferent axons. The immature neurites of multipolar cells closely contact TAG-1-positive axons and generate axons. Inhibition of TAG-1-mediated cell-to-cell interaction or its downstream kinase Lyn impairs neuronal polarization. These results show that the TAG-1-mediated cell-to-cell interaction between the unpolarized multipolar cells and the pioneering axons regulates the polarization of multipolar cells partly through Lyn kinase and Rac1.
Collapse
Affiliation(s)
- Takashi Namba
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Yuji Kibe
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Takuji Ueno
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Akiko Shimada
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Sachi Kozawa
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Mayumi Okamoto
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Yasushi Shimoda
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomiokamachi, Nagaoka, Niigata 940-2188, Japan
| | - Kanako Oda
- Experimental Animal Resource, Brain Research Institute, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahi-machi, Chuo-ku, Niigata, Niigata 951-8510, Japan
| | - Yoshino Wada
- Division of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahi-machi, Chuo-ku, Niigata, Niigata 951-8510, Japan
| | - Tomoyuki Masuda
- Department of Neurobiology, University of Tsukuba School of Medicine, Ibaraki 305-8577, Japan
| | - Akira Sakakibara
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Michihiro Igarashi
- Division of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahi-machi, Chuo-ku, Niigata, Niigata 951-8510, Japan
| | - Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Catherine Faivre-Sarrailh
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, UMR 7286 CNRS, Marseille, France
| | - Kosei Takeuchi
- Division of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahi-machi, Chuo-ku, Niigata, Niigata 951-8510, Japan; Department of Biology, School of Medicine, Aichi Medical University, Yazako, Nagakute, Aichi 480-1195, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan.
| |
Collapse
|
39
|
Abstract
The proper formation and morphogenesis of dendrites is fundamental to the establishment of neural circuits in the brain. Following cell cycle exit and migration, neurons undergo organized stages of dendrite morphogenesis, which include dendritic arbor growth and elaboration followed by retraction and pruning. Although these developmental stages were characterized over a century ago, molecular regulators of dendrite morphogenesis have only recently been defined. In particular, studies in Drosophila and mammalian neurons have identified numerous cell-intrinsic drivers of dendrite morphogenesis that include transcriptional regulators, cytoskeletal and motor proteins, secretory and endocytic pathways, cell cycle-regulated ubiquitin ligases, and components of other signaling cascades. Here, we review cell-intrinsic drivers of dendrite patterning and discuss how the characterization of such crucial regulators advances our understanding of normal brain development and pathogenesis of diverse cognitive disorders.
Collapse
Affiliation(s)
- Sidharth V Puram
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
40
|
Abstract
The ability of injured axons to regenerate declines with age, yet the mechanisms that regulate axon regeneration in response to age are not known. Here we show that axon regeneration in aging C. elegans motor neurons is inhibited by the conserved insulin/IGF1 receptor DAF-2. DAF-2's function in regeneration is mediated by intrinsic neuronal activity of the forkhead transcription factor DAF-16/FOXO. DAF-16 regulates regeneration independently of lifespan, indicating that neuronal aging is an intrinsic, neuron-specific, and genetically regulated process. In addition, we found that DAF-18/PTEN inhibits regeneration independently of age and FOXO signaling via the TOR pathway. Finally, DLK-1, a conserved regulator of regeneration, is downregulated by insulin/IGF1 signaling, bound by DAF-16 in neurons, and required for both DAF-16- and DAF-18-mediated regeneration. Together, our data establish that insulin signaling specifically inhibits regeneration in aging adult neurons and that this mechanism is independent of PTEN and TOR.
Collapse
|
41
|
Estevez AO, Morgan KL, Szewczyk NJ, Gems D, Estevez M. The neurodegenerative effects of selenium are inhibited by FOXO and PINK1/PTEN regulation of insulin/insulin-like growth factor signaling in Caenorhabditis elegans. Neurotoxicology 2014; 41:28-43. [PMID: 24406377 PMCID: PMC3979119 DOI: 10.1016/j.neuro.2013.12.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/23/2013] [Accepted: 12/27/2013] [Indexed: 12/12/2022]
Abstract
Insulin/insulin-like signaling reduction alters selenium-induced neurodegeneration. Selenium induces nuclear translocation of DAF-16/FOXO3a. DAF-16 overexpression decreases GABAergic and cholinergic motor neuron degeneration. Loss of DAF-18/PTEN increases sensitivity to selenium-induced movement deficits. Glutathione requires DAF-18/PINK-1 to improve selenium-induced movement deficits.
Exposures to high levels of environmental selenium have been associated with motor neuron disease in both animals and humans and high levels of selenite have been identified in the cerebrospinal fluid of patients with amyotrophic lateral sclerosis (ALS). We have shown previously that exposures to high levels of sodium selenite in the environment of Caenorhabditis elegans adult animals can induce neurodegeneration and cell loss resulting in motor deficits and death and that this is at least partially caused by a reduction in cholinergic signaling across the neuromuscular junction. Here we provide evidence that reduction in insulin/insulin-like (IIS) signaling alters response to high dose levels of environmental selenium which in turn can regulate the IIS pathway. Most specifically we show that nuclear localization and thus activation of the DAF-16/forkhead box transcription factor occurs in response to selenium exposure although this was not observed in motor neurons of the ventral cord. Yet, tissue specific expression and generalized overexpression of DAF-16 can partially rescue the neurodegenerative and behavioral deficits observed with high dose selenium exposures in not only the cholinergic, but also the GABAergic motor neurons. In addition, two modifiers of IIS signaling, PTEN (phosphatase and tensin homolog, deleted on chromosome 10) and PINK1 (PTEN-induced putative kinase 1) are required for the cellular antioxidant reduced glutathione to mitigate the selenium-induced movement deficits. Studies have suggested that environmental exposures can lead to ALS or other neurological diseases and this model of selenium-induced neurodegeneration developed in a genetically tractable organism provides a tool for examining the combined roles of genetics and environment in the neuro-pathologic disease process.
Collapse
Affiliation(s)
- Annette O Estevez
- Department of Neurology, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| | - Kathleen L Morgan
- Veterans Affairs Pittsburgh Healthcare System, Research and Development (151U), University Drive C, Pittsburgh, PA 15240, USA.
| | - Nathaniel J Szewczyk
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - David Gems
- Institute of Healthy Ageing, and Department of Genetics, Evolution, and Environment, University College London, The Darwin Building, Gower Street, London WC1E 6BT, UK.
| | - Miguel Estevez
- Department of Neurology, University of Arizona College of Medicine, Tucson, AZ 85724, USA; Veterans Affairs Pittsburgh Healthcare System, Research and Development (151U), University Drive C, Pittsburgh, PA 15240, USA.
| |
Collapse
|
42
|
Grossman EN, Giurumescu CA, Chisholm AD. Mechanisms of ephrin receptor protein kinase-independent signaling in amphid axon guidance in Caenorhabditis elegans. Genetics 2013; 195:899-913. [PMID: 23979582 PMCID: PMC3813872 DOI: 10.1534/genetics.113.154393] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 08/16/2013] [Indexed: 12/30/2022] Open
Abstract
Eph receptors and their ephrin ligands are key conserved regulators of axon guidance and can function in a variety of signaling modes. Here we analyze the genetic and cellular requirements for Eph signaling in a Caenorhabditis elegans axon guidance choice point, the ventral guidance of axons in the amphid commissure. The C. elegans Eph receptor EFN-1 has both kinase-dependent and kinase-independent roles in amphid ventral guidance. Of the four C. elegans ephrins, we find that only EFN-1 has a major role in amphid axon ventral guidance, and signals in both a receptor kinase-dependent and kinase-independent manner. Analysis of EFN-1 and EFN-1 expression and tissue-specific requirements is consistent with a model in which VAB-1 acts in amphid neurons, interacting with EFN-1 expressed on surrounding cells. Unexpectedly, left-hand neurons are more strongly affected than right-hand neurons by loss of Eph signaling, indicating a previously undetected left-right asymmetry in the requirement for Eph signaling. By screening candidate genes involved in Eph signaling, we find that the Eph kinase-independent pathway involves the ABL-1 nonreceptor tyrosine kinase and possibly the phosphatidylinositol 3-kinase pathway. Overexpression of ABL-1 is sufficient to rescue EFN-1 ventral guidance defects cell autonomously. Our results reveal new aspects of Eph signaling in a single axon guidance decision in vivo.
Collapse
Affiliation(s)
- Emily N. Grossman
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093
| | - Claudiu A. Giurumescu
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093
| | - Andrew D. Chisholm
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
43
|
Insulin activates the insulin receptor to downregulate the PTEN tumour suppressor. Oncogene 2013; 33:3878-85. [DOI: 10.1038/onc.2013.347] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 07/09/2013] [Accepted: 07/15/2013] [Indexed: 12/26/2022]
|
44
|
Kennedy LM, Pham SCDL, Grishok A. Nonautonomous regulation of neuronal migration by insulin signaling, DAF-16/FOXO, and PAK-1. Cell Rep 2013; 4:996-1009. [PMID: 23994474 DOI: 10.1016/j.celrep.2013.07.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 07/02/2013] [Accepted: 07/30/2013] [Indexed: 01/29/2023] Open
Abstract
Neuronal migration is essential for nervous system development in all organisms and is regulated in the nematode, C. elegans, by signaling pathways that are conserved in humans. Here, we demonstrate that the insulin/IGF-1-PI3K signaling pathway modulates the activity of the DAF-16/FOXO transcription factor to regulate the anterior migrations of the hermaphrodite-specific neurons (HSNs) during embryogenesis of C. elegans. When signaling is reduced, DAF-16 is activated and promotes migration; conversely, when signaling is enhanced, DAF-16 is inactivated, and migration is inhibited. We show that DAF-16 acts nonautonomously in the hypodermis to promote HSN migration. Furthermore, we identify PAK-1, a p21-activated kinase, as a downstream mediator of insulin/IGF-1-DAF-16 signaling in the nonautonomous control of HSN migration. Because a FOXO-Pak1 pathway was recently shown to regulate mammalian neuronal polarity, our findings indicate that the roles of FOXO and Pak1 in neuronal migration are most likely conserved from C. elegans to higher organisms.
Collapse
Affiliation(s)
- Lisa M Kennedy
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | | | | |
Collapse
|
45
|
Parmentier F, Lejeune FX, Neri C. Pathways to decoding the clinical potential of stress response FOXO-interaction networks for Huntington's disease: of gene prioritization and context dependence. Front Aging Neurosci 2013; 5:22. [PMID: 23781200 PMCID: PMC3680703 DOI: 10.3389/fnagi.2013.00022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 05/27/2013] [Indexed: 01/06/2023] Open
Abstract
The FOXO family of transcription factors is central to the regulation of organismal longevity and cellular survival. Several studies have indicated that FOXO factors lie at the center of a complex network of upstream pathways, cofactors and downstream targets (FOXO-interaction networks), which may have developmental and post-developmental roles in the regulation of chronic-stress response in normal and diseased cells. Noticeably, FOXO factors are important for the regulation of proteotoxicity and neuron survival in several models of neurodegenerative disease, suggesting that FOXO-interaction networks may have therapeutic potential. However, the status of FOXO-interaction networks in neurodegenerative disease remains largely unknown. Systems modeling is anticipated to provide a comprehensive assessment of this question. In particular, interrogating the context-dependent variability of FOXO-interaction networks could predict the clinical potential of cellular-stress response genes and aging regulators for tackling brain and peripheral pathology in neurodegenerative disease. Using published transcriptomic data obtained from murine models of Huntington's disease (HD) and post-mortem brains, blood samples and induced-pluripotent-stem cells from HD carriers as a case example, this review briefly highlights how the biological status and clinical potential of FOXO-interaction networks for HD may be decoded by developing network and entropy based feature selection across heterogeneous datasets.
Collapse
Affiliation(s)
- Frédéric Parmentier
- Laboratory of Neuronal Cell biology and Pathology, INSERM Unit 894, CNRS UMR 7102, University Pierre and Marie Curie Paris, France
| | | | | |
Collapse
|
46
|
Park JH, Lee SB, Lee KH, Ahn JY. Nuclear Akt promotes neurite outgrowth in the early stage of neuritogenesis. BMB Rep 2013; 45:521-5. [PMID: 23010173 DOI: 10.5483/bmbrep.2012.45.9.114] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In addition to its pivotal role in neuronal survival, PI3K/Akt signaling is integral to neuronal differentiation and neurite outgrowth. However, the exact role of Akt in neuronal differentiation is still controversial. Here, we found that nuclear expression of CA-Akt resulted in unusual rapid neurite outgrowth and overexpression of KD-Akt caused multiple dendrite growth without specific axon elongation. Moreover, microarray data revealed that the expression of FOXQ1 expression was about 10-fold higher in cells with nuclear, active Akt than in control cells. Quantitative real-time PCR analysis showed that mRNA levels were upregulated in NLS-CA-Akt cells as compared to KD or EV cells. Furthermore, our FACS analysis demonstrated that overexpression of NLS-CA-Akt accumulate cells in the G1 phase within 24 h, fitting with the rapid sprouting of neuritis. Thus, our data implied that at least in this early time frame, the overexpression of nuclear, active Akt forced cells into neurite development through probably FOXQ1regulation.
Collapse
Affiliation(s)
- Ji Hye Park
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea
| | | | | | | |
Collapse
|
47
|
Nakdimon I, Walser M, Fröhli E, Hajnal A. PTEN negatively regulates MAPK signaling during Caenorhabditis elegans vulval development. PLoS Genet 2012; 8:e1002881. [PMID: 22916028 PMCID: PMC3420937 DOI: 10.1371/journal.pgen.1002881] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 06/19/2012] [Indexed: 11/20/2022] Open
Abstract
Vulval development in Caenorhabditis elegans serves as an excellent model to examine the crosstalk between different conserved signaling pathways that are deregulated in human cancer. The concerted action of the RAS/MAPK, NOTCH, and WNT pathways determines an invariant pattern of cell fates in three vulval precursor cells. We have discovered a novel form of crosstalk between components of the Insulin and the RAS/MAPK pathways. The insulin receptor DAF-2 stimulates, while DAF-18 PTEN inhibits, RAS/MAPK signaling in the vulval precursor cells. Surprisingly, the inhibitory activity of DAF-18 PTEN on the RAS/MAPK pathway is partially independent of its PIP3 lipid phosphatase activity and does not involve further downstream components of the insulin pathway, such as AKT and DAF-16 FOXO. Genetic and biochemical analyses indicate that DAF-18 negatively regulates vulval induction by inhibiting MAPK activation. Thus, mutations in the PTEN tumor suppressor gene may result in the simultaneous hyper-activation of two oncogenic signaling pathways. The human tumor suppressor PTEN is mutated in many different types of cancer. Using the roundworm C. elegans as a model to study how cells communicate during animal development, we discovered a new mechanism by which PTEN inhibits the activity of the oncogenic RAS/MAPK signaling pathway. Focusing on the development of the vulva, the egg-laying organ of the hermaphrodite, as a model to investigate the regulation of RAS/MAPK signaling, we could distinguish between two distinct inhibitory activities of PTEN on the RAS/MAPK signaling pathway. On the one hand, PTEN acts as a lipid phosphatase that inhibits the production of PIP3, which in turn stimulates RAS/MAPK signaling. On the other hand, PTEN acts as a protein phosphatase that negatively regulates RAS/MAPK signaling by inhibiting signal transduction at the level of the MAPK, which is a key component in the pathway. Understanding the detailed molecular mechanism by which the PTEN tumor suppressor homolog regulates signal transduction in C. elegans can help predict the consequences of mutations in human PTEN for cancer development in humans.
Collapse
Affiliation(s)
- Itay Nakdimon
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Cancer Network Zürich PhD Program, Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Michael Walser
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Sciences PhD Program, Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Erika Fröhli
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Alex Hajnal
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
48
|
Ostrakhovitch EA, Semenikhin OA. The role of redox environment in neurogenic development. Arch Biochem Biophys 2012; 534:44-54. [PMID: 22910298 DOI: 10.1016/j.abb.2012.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/19/2012] [Accepted: 08/03/2012] [Indexed: 10/28/2022]
Abstract
The dynamic changes of cellular redox elements during neurogenesis allow the control of specific programs for selective lineage progression. There are many redox couples that influence the cellular redox state. The shift from a reduced to an oxidized state and vice versa may act as a cellular switch mechanism of stem cell mode of action from proliferation to differentiation. The redox homeostasis ensures proper functioning of redox-sensitive signaling pathways through oxidation/reduction of critical cysteine residues on proteins involved in signal transduction. This review presents the current knowledge on the relation between changes in the cellular redox environment and stem cell programming in the course of commitment to a restricted neural lineage, focusing on in vivo neurogenesis and in vitro neuronal differentiation. The first two sections outline the main systems that control the intracellular redox environment and make it more oxidative or reductive. The last section provides the background on redox-sensitive signaling pathways that regulate neurogenesis.
Collapse
Affiliation(s)
- E A Ostrakhovitch
- Department of Chemistry, The University of Western Ontario, London, Ontario, Canada N6A 5B7.
| | | |
Collapse
|
49
|
Nechipurenko IV, Broihier HT. FoxO limits microtubule stability and is itself negatively regulated by microtubule disruption. ACTA ACUST UNITED AC 2012; 196:345-62. [PMID: 22312004 PMCID: PMC3275378 DOI: 10.1083/jcb.201105154] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
FoxO inhibits microtubule stability in the central nervous system, making its degradation an essential component of a cell’s protective response to cytoskeletal insult. Transcription factors are essential for regulating neuronal microtubules (MTs) during development and after axon damage. In this paper, we identify a novel neuronal function for Drosophila melanogaster FoxO in limiting MT stability at the neuromuscular junction (NMJ). foxO loss-of-function NMJs displayed augmented MT stability. In contrast, motor neuronal overexpression of wild-type FoxO moderately destabilized MTs, whereas overexpression of constitutively nuclear FoxO severely destabilized MTs. Thus, FoxO negatively regulates synaptic MT stability. FoxO family members are well-established components of stress-activated feedback loops. We hypothesized that FoxO might also be regulated by cytoskeletal stress because it was well situated to shape neuronal MT organization after cytoskeletal damage. Indeed, levels of neuronal FoxO were strongly reduced after acute pharmacological MT disruption as well as sustained genetic disruption of the neuronal cytoskeleton. This decrease was independent of the dual leucine zipper kinase–Wallenda pathway and required function of Akt kinase. We present a model wherein FoxO degradation is a component of a stabilizing, protective response to cytoskeletal insult.
Collapse
Affiliation(s)
- Inna V Nechipurenko
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | | |
Collapse
|