1
|
Ragle JM, Turzo A, Jackson A, Vo AA, Pham VT, Ward JD. The NHR-23-regulated putative protease inhibitor mlt-11 gene is necessary for C. elegans cuticle structure and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.12.593762. [PMID: 38766248 PMCID: PMC11100798 DOI: 10.1101/2024.05.12.593762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
C. elegans molting offers a powerful entry point to understanding developmentally programmed apical extracellular matrix remodeling. However, the gene regulatory network controlling this process remains poorly understood. Focusing on targets of NHR-23, a key transcription factor that drives molting, we confirmed the Kunitz family protease inhibitor gene mlt-11 as an NHR-23 target. Through reporter assays, we identified NHR-23-binding sites that are necessary and sufficient for epithelial expression. We generated a translational fusion and demonstrated that MLT-11 is localized to the cuticle and lined openings to the exterior (vulva, rectum, mouth). We created a set of strains expressing varied levels of MLT-11 by deleting endogenous cis-regulatory element sequences. Combined deletion of two cis-regulatory elements caused developmental delay, motility defects, and failure of the cuticle barrier. Inactivation of mlt-11 by RNAi produced even more pronounced defects. mlt-11 is necessary to pattern every layer of the adult cuticle, suggesting a broad patterning role prior to the formation of the mature cuticle. Together these studies provide an entry point into understanding how individual cis-regulatory elements function to coordinate expression of oscillating genes involved in molting and how MLT-11 ensures proper cuticle assembly.
Collapse
Affiliation(s)
- James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Ariela Turzo
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Anton Jackson
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - An A. Vo
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Vivian T. Pham
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jordan D. Ward
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
2
|
Hernandez-Lima MA, Seo B, Urban ND, Truttmann MC. C. elegans behavior, fitness, and lifespan, are modulated by AWB/ASH-dependent death perception. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617097. [PMID: 39416137 PMCID: PMC11482816 DOI: 10.1101/2024.10.07.617097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The ability of the nervous system to initiate intricate goal-directed behaviors in response to environmental stimuli is essential for metazoan survival. In this study, we demonstrate that the nematode Caenorhabditis elegans perceives and reacts to dead conspecifics. The exposure to C. elegans corpses as well as corpse lysates activates sensory neurons AWB and ASH, triggering a glutamate- and acetylcholine-dependent signaling cascade that regulates both immediate (aversion) and long-term (survival) responses to the presence of a death signature. We identify increased adenosine monophosphate (AMP) and cysteine concentrations as chemical fingerprints for the presence of metazoan corpses and show that death cue sensing by AWB and ASH leads to physiological changes which promote reproduction at the expense of lifespan. Our findings illuminate a novel signaling paradigm that allows organisms to detect and interpret the environmental enrichment of intracellular metabolites as a death cue.
Collapse
Affiliation(s)
- Mirella A. Hernandez-Lima
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Brian Seo
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nicholas D. Urban
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Graduate Program in Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthias C. Truttmann
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Graduate Program in Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Geriatrics Center, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
3
|
Guo A, Wu Q, Yan X, Chen K, Liu Y, Liang D, Yang Y, Luo Q, Xiong M, Yu Y, Fei E, Chen F. Differential roles of lysosomal cholesterol transporters in the development of C. elegans NMJs. Life Sci Alliance 2024; 7:e202402584. [PMID: 39084875 PMCID: PMC11291935 DOI: 10.26508/lsa.202402584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Cholesterol homeostasis in neurons is critical for synapse formation and maintenance. Neurons with impaired cholesterol uptake undergo progressive synapse loss and eventual degeneration. To investigate the molecular mechanisms of neuronal cholesterol homeostasis and its role during synapse development, we studied motor neurons of Caenorhabditis elegans because these neurons rely on dietary cholesterol. Combining lipidomic analysis, we discovered that NCR-1, a lysosomal cholesterol transporter, promotes cholesterol absorption and synapse development. Loss of ncr-1 causes smaller synapses, and low cholesterol exacerbates the deficits. Moreover, NCR-1 deficiency hinders the increase in synapses under high cholesterol. Unexpectedly, NCR-2, the NCR-1 homolog, increases the use of cholesterol and sphingomyelins and impedes synapse formation. NCR-2 deficiency causes an increase in synapses regardless of cholesterol concentration. Inhibiting the degradation or synthesis of sphingomyelins can induce or suppress the synaptic phenotypes in ncr-2 mutants. Our findings indicate that neuronal cholesterol homeostasis is differentially controlled by two lysosomal cholesterol transporters and highlight the importance of neuronal cholesterol homeostasis in synapse development.
Collapse
Affiliation(s)
- Amin Guo
- https://ror.org/042v6xz23 School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qi Wu
- https://ror.org/042v6xz23 School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xin Yan
- https://ror.org/042v6xz23 School of Life Sciences, Nanchang University, Nanchang, China
| | - Kanghua Chen
- https://ror.org/042v6xz23 School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuxiang Liu
- https://ror.org/042v6xz23 School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dingfa Liang
- https://ror.org/042v6xz23 Queen Mary School of Nanchang University, Jiangxi Medical College, Nanchang, China
| | - Yuxiao Yang
- https://ror.org/042v6xz23 School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qunfeng Luo
- https://ror.org/042v6xz23 School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Mingtao Xiong
- https://ror.org/042v6xz23 Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Erkang Fei
- https://ror.org/042v6xz23 Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fei Chen
- https://ror.org/042v6xz23 School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Gordon SG, Rodriguez AA, Gu Y, Corbett KD, Lee CF, Rog O. The synaptonemal complex aligns meiotic chromosomes by wetting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.07.607092. [PMID: 39149313 PMCID: PMC11326210 DOI: 10.1101/2024.08.07.607092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
During meiosis, the parental chromosomes are drawn together to enable exchange of genetic information. Chromosomes are aligned through the assembly of a conserved interface, the synaptonemal complex, composed of a central region that forms between two parallel chromosomal backbones called axes. Here we identify the axis-central region interface in C. elegans, containing a conserved positive patch on the axis component HIM-3 and the C-terminus of the central region protein SYP-5. Crucially, the canonical ultrastructure of the synaptonemal complex is altered upon weakening this interface. We developed a thermodynamic model that recapitulates our experimental observations, indicating that the liquid-like central region can assemble by wetting the axes without active energy consumption. More broadly, our data show that condensation drives tightly regulated nuclear reorganization during sexual reproduction.
Collapse
Affiliation(s)
- Spencer G. Gordon
- School of Biological Sciences and Center for Cell and Genome Sciences, University of Utah, United States
| | - Alyssa A. Rodriguez
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla CA 92093
| | - Yajie Gu
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla CA 92093
| | - Kevin D. Corbett
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla CA 92093
- Department of Molecular Biology, University of California San Diego, La Jolla CA 92093
| | - Chiu Fan Lee
- Department of Bioengineering, Imperial College London, United Kingdom
| | - Ofer Rog
- School of Biological Sciences and Center for Cell and Genome Sciences, University of Utah, United States
| |
Collapse
|
5
|
Zhang P, Medwig-Kinney TN, Breiner EA, Perez JM, Song AN, Goldstein B. Cell signaling facilitates apical constriction by basolaterally recruiting Arp2/3 via Rac and WAVE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614059. [PMID: 39386716 PMCID: PMC11463545 DOI: 10.1101/2024.09.23.614059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Apical constriction is a critical cell shape change that bends tissues. How precisely-localized actomyosin regulators drive apical constriction remains poorly understood. C. elegans gastrulation provides a valuable model to address this question. The Arp2/3 complex is essential in C. elegans gastrulation. To understand how Arp2/3 is locally regulated, we imaged embryos with endogenously-tagged Arp2/3 and its nucleation-promoting factors (NPFs). The three NPFs - WAVE, WASP, and WASH - colocalized with Arp2/3 and controlled Arp2/3 localization at distinct subcellular locations. We exploited this finding to study distinct populations of Arp2/3 and found that only WAVE depletion caused penetrant gastrulation defects. WAVE localized basolaterally with Arp2/3 at cell-cell contacts, dependent on CED-10/Rac. Establishing ectopic cell contacts recruited WAVE and Arp2/3, identifying contact as a symmetry-breaking cue for localization of these proteins. These results suggest that cell-cell signaling via Rac activates WAVE and Arp2/3 basolaterally, and that basolateral Arp2/3 is important for apical constriction.
Collapse
Affiliation(s)
- Pu Zhang
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Eleanor A. Breiner
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jadyn M. Perez
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - April N. Song
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bob Goldstein
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
6
|
Ranjan R, Ma B, Gleason RJ, Liao Y, Bi Y, Davis BEM, Yang G, Clark M, Mahajan V, Condon M, Broderick NA, Chen X. Modulating DNA Polα Enhances Cell Reprogramming Across Species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613993. [PMID: 39345551 PMCID: PMC11429986 DOI: 10.1101/2024.09.19.613993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
As a fundamental biological process, DNA replication ensures the accurate copying of genetic information. However, the impact of this process on cellular plasticity in multicellular organisms remains elusive. Here, we find that reducing the level or activity of a replication component, DNA Polymerase α (Polα), facilitates cell reprogramming in diverse stem cell systems across species. In Drosophila male and female germline stem cell lineages, reducing Polα levels using heterozygotes significantly enhances fertility of both sexes, promoting reproductivity during aging without compromising their longevity. Consistently, in C. elegans the pola heterozygous hermaphrodites exhibit increased fertility without a reduction in lifespan, suggesting that this phenomenon is conserved. Moreover, in male germline and female intestinal stem cell lineages of Drosophila, polα heterozygotes exhibit increased resistance to tissue damage caused by genetic ablation or pathogen infection, leading to enhanced regeneration and improved survival during post-injury recovery, respectively. Additionally, fine tuning of an inhibitor to modulate Polα activity significantly enhances the efficiency of reprogramming human embryonic fibroblasts into induced pluripotent cells. Together, these findings unveil novel roles of a DNA replication component in regulating cellular reprogramming potential, and thus hold promise for promoting tissue health, facilitating post-injury rehabilitation, and enhancing healthspan.
Collapse
Affiliation(s)
- Rajesh Ranjan
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| | - Binbin Ma
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| | - Ryan J. Gleason
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yijun Liao
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yingshan Bi
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brendon E. M. Davis
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Guanghui Yang
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| | - Maggie Clark
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Vikrant Mahajan
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Madison Condon
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Xin Chen
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| |
Collapse
|
7
|
Moseley-Alldredge M, Aragón C, Vargus M, Alley D, Somia N, Chen L. The L1CAM SAX-7 is an antagonistic modulator of Erk Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.14.613091. [PMID: 39345534 PMCID: PMC11429911 DOI: 10.1101/2024.09.14.613091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
L1CAMs are immunoglobulin superfamily cell adhesion molecules that ensure proper nervous system development and function. In addition to being associated with the autism and schizophrenia spectrum disorders, mutations in the L1CAM family of genes also underlie distinct developmental syndromes with neurological conditions, such as intellectual disability, spastic paraplegia, hypotonia and congenital hydrocephalus. Studies in both vertebrate and invertebrate model organisms have established conserved neurodevelopmental roles for L1CAMs; these include axon guidance, dendrite morphogenesis, synaptogenesis, and maintenance of neural architecture, among others. In Caenorhabditis elegans , L1CAMs, encoded by the sax-7 gene, are required for coordinated locomotion. We previously uncovered a genetic interaction between sax-7 and components of synaptic vesicle cycle, revealing a non-developmental role for sax-7 in regulating synaptic activity. More recently, we determined that sax-7 also genetically interacts with extracellular signal-related kinase (ERK) signaling in controlling coordinated locomotion. C. elegans ERK, encoded by the mpk-1 gene, is a serine/threonine protein kinase belonging to the mitogen-activated protein kinase (MAPK) family that governs multiple aspects of animal development and cellular homeostasis. Here, we show this genetic interaction between sax-7 and mpk-1 occurs not only in cholinergic neurons for coordinated locomotion, but also extends outside the nervous system, revealing novel roles for SAX-7/L1CAM in non-neuronal processes, including vulval development. Our genetic findings in both the nervous system and developing vulva are consistent with SAX-7/L1CAM acting as an antagonistic modulator of ERK signaling.
Collapse
|
8
|
Ananthaswamy D, Funes K, Borges T, Roques S, Fassnacht N, Jamal SE, Checchi PM, Wei-sy Lee T. NuRD chromatin remodeling is required to repair exogenous DSBs in the Caenorhabditis elegans germline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.14.613027. [PMID: 39314477 PMCID: PMC11419128 DOI: 10.1101/2024.09.14.613027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Organisms rely on coordinated networks of DNA repair pathways to protect genomes against toxic double-strand breaks (DSBs), particularly in germ cells. All repair mechanisms must successfully negotiate the local chromatin environment in order to access DNA. For example, nucleosomes can be repositioned by the highly conserved Nucleosome Remodeling and Deacetylase (NuRD) complex. In Caenorhabditis elegans, NuRD functions in the germline to repair DSBs - the loss of NuRD's ATPase subunit, LET-418/CHD4, prevents DSB resolution and therefore reduces fertility. In this study, we challenge germlines with exogenous DNA damage to better understand NuRD's role in repairing DSBs. We find that let-418 mutants are hypersensitive to cisplatin and hydroxyurea: exposure to either mutagen impedes DSB repair, generates aneuploid oocytes, and severely reduces fertility and embryonic survival. These defects resemble those seen when the Fanconi anemia (FA) DNA repair pathway is compromised, and we find that LET-418's activity is epistatic to that of the FA component FCD-2/FANCD2. We propose a model in which NuRD is recruited to the site of DNA lesions to remodel chromatin and allow access for FA pathway components. Together, these results implicate NuRD in the repair of both endogenous DSBs and exogenous DNA lesions to preserve genome integrity in developing germ cells.
Collapse
Affiliation(s)
- Deepshikha Ananthaswamy
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Dr. Lowell MA, 01854
| | - Kelin Funes
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Dr. Lowell MA, 01854
| | - Thiago Borges
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Dr. Lowell MA, 01854
| | - Scott Roques
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Dr. Lowell MA, 01854
| | - Nina Fassnacht
- Department of Biology, Marist College, 3399 North Road, Poughkeepsie, NY 12601
| | - Sereen El Jamal
- Department of Biology, Marist College, 3399 North Road, Poughkeepsie, NY 12601
| | - Paula M. Checchi
- Department of Biology, Marist College, 3399 North Road, Poughkeepsie, NY 12601
| | - Teresa Wei-sy Lee
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Dr. Lowell MA, 01854
| |
Collapse
|
9
|
Mialon M, Patrash L, Weinreb A, Özkan E, Bessereau JL, Pinan-Lucarre B. A trans-synaptic IgLON adhesion molecular complex directly contacts and clusters a nicotinic receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611427. [PMID: 39314492 PMCID: PMC11418930 DOI: 10.1101/2024.09.05.611427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The localization and clustering of neurotransmitter receptors at appropriate postsynaptic sites is a key step in the control of synaptic transmission. Here, we identify a novel paradigm for the synaptic localization of an ionotropic acetylcholine receptor (AChR) based on the direct interaction of its extracellular domain with a cell adhesion molecule of the IgLON family. Our results show that RIG-5 and ZIG-8, which encode the sole IgLONs in C. elegans, are tethered in the pre- and postsynaptic membranes, respectively, and interact in vivo through their first immunoglobulin-like (Ig) domains. In addition, ZIG-8 traps ACR-16 via a direct cis- interaction between the ZIG-8 Ig2 domain and the base of the large extracellular AChR domain. Such mechanism has never been reported, but all these molecules are conserved during evolution. Similar interactions may directly couple Ig superfamily adhesion molecules and members of the large family of Cys-loop ionotropic receptors, including AChRs, in the mammalian nervous system, and may be relevant in the context of IgLON-associated brain diseases.
Collapse
|
10
|
Kasimatis KR, Willis JH, Sedore CA, Phillips PC. Transcriptomic sexual conflict at two evolutionary timescales revealed by experimental evolution in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.09.552689. [PMID: 37609247 PMCID: PMC10441408 DOI: 10.1101/2023.08.09.552689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Sex-specific regulation of gene expression is the most plausible way for generating sexually differentiated phenotypes from an essentially shared genome. However, since genetic material is shared, sex-specific selection in one sex can have an indirect response in the other sex. From a gene expression perspective, this tethered response can move one sex away from their wildtype expression state and impact potentially many gene regulatory networks. Here, using experimental evolution in the model nematode Caenorhabditis elegans, we explore the coupling of direct sexual selection on males with the transcriptomic response in males and females over microevolutionary timescales to uncover the extent to which post-insemination reproductive traits share a genetic basis between the sexes. We find that differential gene expression evolved in a sex-specific manner in males, while in females indirect selection causes an evolved response. Almost all differentially expressed genes were downregulated in both evolved males and females. Moreover, 97% of significantly differentially expressed genes in males and 69% of significantly differentially expressed genes in females have wildtype female-biased expression profile. Changes in gene expression profiles were likely driven through trans -acting pathways that are shared between the sexes. We found no evidence that the core dosage compensation machinery was impacted by experimental evolution. Together these data suggest a de-feminization of the male transcriptome and masculinization of the female transcriptome driven by direct selection on male sperm competitive ability. Our results indicate that on short evolutionary timescales sexual selection can generate putative sexual conflict in expression space.
Collapse
|
11
|
Ahmed M, Fischer S, Robert KL, Lange KI, Stuck MW, Best S, Johnson CA, Pazour GJ, Blacque OE, Nandadasa S. Two functional forms of the Meckel-Gruber syndrome protein TMEM67 generated by proteolytic cleavage by ADAMTS9 mediate Wnt signaling and ciliogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611229. [PMID: 39282264 PMCID: PMC11398388 DOI: 10.1101/2024.09.04.611229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
TMEM67 mutations are the major cause of Meckel-Gruber syndrome. TMEM67 is involved in both ciliary transition zone assembly, and non-canonical Wnt signaling mediated by its extracellular domain. How TMEM67 performs these two separate functions is not known. We identify a novel cleavage motif in the extracellular domain of TMEM67 cleaved by the extracellular matrix metalloproteinase ADAMTS9. This cleavage regulates the abundance of two functional forms: A C-terminal portion which localizes to the ciliary transition zone regulating ciliogenesis, and a non-cleaved form which regulates Wnt signaling. By characterizing three TMEM67 ciliopathy patient variants within the cleavage motif utilizing mammalian cell culture and C. elegans, we show the cleavage motif is essential for cilia structure and function, highlighting its clinical significance. We generated a novel non-cleavable TMEM67 mouse model which develop severe ciliopathies phenocopying Tmem67 -/- mice, but in contrast, undergo normal Wnt signaling, substantiating the existence of two functional forms of TMEM67.
Collapse
Affiliation(s)
- Manu Ahmed
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA
| | - Sydney Fischer
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA
| | - Karyn L. Robert
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA
| | - Karen I. Lange
- School of Biomolecular & Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Michael W. Stuck
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA
| | - Sunayna Best
- Division of Molecular Medicine, Leeds Institute of Medical Research, The University of Leeds, Leeds, UK
- Department of Clinical Genetics, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Colin A. Johnson
- Division of Molecular Medicine, Leeds Institute of Medical Research, The University of Leeds, Leeds, UK
| | - Gregory J. Pazour
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA
| | - Oliver E. Blacque
- School of Biomolecular & Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sumeda Nandadasa
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA
| |
Collapse
|
12
|
Mackie M, Le VV, Carstensen HR, Kushnir NR, Castro DL, Dimov IM, Quach KT, Cook SJ, Hobert O, Chalasani SH, Hong RL. Evolution of lateralized gustation in nematodes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.31.610597. [PMID: 39282255 PMCID: PMC11398344 DOI: 10.1101/2024.08.31.610597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Animals with small nervous systems have a limited number of sensory neurons that must encode information from a changing environment. This problem is particularly exacerbated in nematodes that populate a wide variety of distinct ecological niches but only have a few sensory neurons available to encode multiple modalities. How does sensory diversity prevail within this neuronal constraint? To identify the genetic basis for patterning different nervous systems, we demonstrate that sensory neurons in the Pristionchus pacificus respond to various salt sensory cues in a manner that is partially distinct from that of the distantly related nematode C. elegans. By visualizing neuronal activity patterns, we show that contrary to previous expectations based on its genome sequence, the salt responses of P. pacificus are encoded in a left/right asymmetric manner in the bilateral ASE neuron pair. Our study illustrates patterns of evolutionary stability and change in the gustatory system of nematodes.
Collapse
Affiliation(s)
- Marisa Mackie
- Department of Biology California State University, Northridge, CA, USA
| | - Vivian Vy Le
- Department of Biology California State University, Northridge, CA, USA
| | | | - Nicole R. Kushnir
- Department of Biology California State University, Northridge, CA, USA
| | - Dylan L. Castro
- Department of Biology California State University, Northridge, CA, USA
| | - Ivan M. Dimov
- Department of Biology California State University, Northridge, CA, USA
| | - Kathleen T. Quach
- Molecular Neurobiology Laboratory Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Steven J. Cook
- Department of Biological Sciences Howard Hughes Medical Institute, Columbia University, New York, NY, USA
- Present address: Neural Coding Department Allen Institute for Brain Science, Seattle, WA, USA
| | - Oliver Hobert
- Department of Biological Sciences Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| | - Sreekanth H. Chalasani
- Molecular Neurobiology Laboratory Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ray L. Hong
- Department of Biology California State University, Northridge, CA, USA
| |
Collapse
|
13
|
Blazie SM, Fortunati D, Zhao Y, Jin Y. C. elegans LIN-66 mediates EIF-3/eIF3-dependent protein translation via a cold-shock domain. Life Sci Alliance 2024; 7:e202402673. [PMID: 38886018 PMCID: PMC11184513 DOI: 10.26508/lsa.202402673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Protein translation initiation is a conserved process involving many proteins acting in concert. The 13 subunit eukaryotic initiation factor 3 (eIF3) complex is essential for assembly of the pre-initiation complex that scans mRNA and positions ribosome at the initiation codon. We previously reported that a gain-of-function (gf) mutation affecting the G subunit of the Caenorhabditis elegans eIF3 complex, eif-3.g(gf), selectively modulates protein translation in the ventral cord cholinergic motor neurons. Here, through unbiased genetic suppressor screening, we identified that the gene lin-66 mediates eif-3.g(gf)-dependent protein translation in motor neurons. LIN-66 is composed largely of low-complexity amino acid sequences with unknown functional domains. We combined bioinformatics analysis with in vivo functional dissection and identified a cold-shock domain in LIN-66 critical for its function. In cholinergic motor neurons, LIN-66 shows a close association with EIF-3.G in the cytoplasm. The low-complexity amino acid sequences of LIN-66 modulate its subcellular pattern. As cold-shock domains function broadly in RNA regulation, we propose that LIN-66 mediates stimulus-dependent protein translation by facilitating the interaction of mRNAs with EIF-3.G.
Collapse
Affiliation(s)
- Stephen M Blazie
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Daniel Fortunati
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yan Zhao
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
14
|
Perry JA, Werner ME, Omi S, Heck BW, Maddox PS, Mavrakis M, Maddox AS. Animal septins contain functional transmembrane domains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.20.567915. [PMID: 38045322 PMCID: PMC10690161 DOI: 10.1101/2023.11.20.567915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Septins, a conserved family of filament-forming proteins, contribute to eukaryotic cell division, polarity, and membrane trafficking. Septins scaffold other proteins to cellular membranes, but it is unknown how septins associate with membranes. We identified and characterized an isoform of Caenorhabditis elegans septin UNC-61 that was predicted to contain a transmembrane domain (TMD). The TMD isoform is expressed in a subset of tissues where the known septins were known to act, and TMD function was required for tissue integrity of the egg-laying apparatus. We found TMD-containing septins across opisthokont phylogeny and demonstrated that the TMD-containing sequence of a primate TMD-septin is sufficient for localization to cellular membranes. Together, our findings reveal a novel mechanism of septin-membrane association with profound implications for septin dynamics and regulation.
Collapse
Affiliation(s)
- Jenna A. Perry
- Department of Biology, The University of North Carolina at Chapel Hill; Chapel Hill, North Carolina, 27599 USA
| | - Michael E. Werner
- Department of Biology, The University of North Carolina at Chapel Hill; Chapel Hill, North Carolina, 27599 USA
| | - Shizue Omi
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Bryan W. Heck
- Department of Biology, The University of North Carolina at Chapel Hill; Chapel Hill, North Carolina, 27599 USA
| | - Paul S. Maddox
- Department of Biology, The University of North Carolina at Chapel Hill; Chapel Hill, North Carolina, 27599 USA
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Amy S. Maddox
- Department of Biology, The University of North Carolina at Chapel Hill; Chapel Hill, North Carolina, 27599 USA
| |
Collapse
|
15
|
Iannacone MJ, Um P, Grubbs JI, van der Linden AM, Raizen DM. Quiescence Enhances Survival during Viral Infection in Caenorhabditis elegans. J Neurosci 2024; 44:e1700222024. [PMID: 39060176 PMCID: PMC11358607 DOI: 10.1523/jneurosci.1700-22.2024] [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: 09/07/2022] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Infection causes reduced activity, anorexia, and sleep, which are components of the phylogenetically conserved but poorly understood sickness behavior. We developed a Caenorhabditis elegans model to study quiescence during chronic infection, using infection with the Orsay virus. The Orsay virus infects intestinal cells yet strongly affects behavior, indicating gut-to-nervous system communication. Infection quiescence has the sleep properties of reduced responsiveness and rapid reversibility. Both the ALA and RIS neurons regulate virus-induced quiescence though ALA plays a more prominent role. Quiescence-defective animals have decreased survival when infected, indicating a benefit of quiescence during chronic infectious disease. The survival benefit of quiescence is not explained by a difference in viral load, indicating that it improves resilience rather than resistance to infection. Orsay infection is associated with a decrease in ATP levels, and this decrease is more severe in quiescence-defective animals. We propose that quiescence preserves energetic resources by reducing energy expenditures and/or by increasing extraction of energy from nutrients. This model presents an opportunity to explore the role of sleep and fatigue in chronic infectious illness.
Collapse
Affiliation(s)
- Michael J Iannacone
- Department of Neurology, and Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Paul Um
- Department of Neurology, and Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Jeremy I Grubbs
- Department of Biology, University of Nevada, Reno, Nevada 89557
| | | | - David M Raizen
- Department of Neurology, and Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
16
|
Lang C, Maxian O, Anneken A, Munro E. Oligomerization and positive feedback on membrane recruitment encode dynamically stable PAR-3 asymmetries in the C. elegans zygote. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.04.552031. [PMID: 39253498 PMCID: PMC11383301 DOI: 10.1101/2023.08.04.552031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Studies of PAR polarity have emphasized a paradigm in which mutually antagonistic PAR proteins form complementary polar domains in response to transient cues. A growing body of work suggests that the oligomeric scaffold PAR-3 can form unipolar asymmetries without mutual antagonism, but how it does so is largely unknown. Here we combine single molecule analysis and modeling to show how the interplay of two positive feedback loops promote dynamically stable unipolar PAR-3 asymmetries in early C. elegans embryos. First, the intrinsic dynamics of PAR-3 membrane binding and oligomerization encode negative feedback on PAR-3 dissociation. Second, membrane-bound PAR-3 promotes its own recruitment through a mechanism that requires the anterior polarity proteins CDC-42, PAR-6 and PKC-3. Using a kinetic model tightly constrained by our experimental measurements, we show that these two feedback loops are individually required and jointly sufficient to encode dynamically stable and locally inducible unipolar PAR-3 asymmetries in the absence of posterior inhibition. Given the central role of PAR-3, and the conservation of PAR-3 membrane-binding, oligomerization, and core interactions with PAR-6/aPKC, these results have widespread implications for PAR-mediated polarity in metazoa.
Collapse
Affiliation(s)
- Charlie Lang
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL 60637
- Current address: Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305
| | - Ondrej Maxian
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Alexander Anneken
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Edwin Munro
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
17
|
Ji H, Chen D, Fang-Yen C. Automated multimodal imaging of Caenorhabditis elegans behavior in multi-well plates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579675. [PMID: 38405855 PMCID: PMC10888940 DOI: 10.1101/2024.02.09.579675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Assays of behavior in model organisms play an important role in genetic screens, drug testing, and the elucidation of gene-behavior relationships. We have developed an automated, high-throughput imaging and analysis method for assaying behaviors of the nematode C. elegans . We use high-resolution optical imaging to longitudinally record the behaviors of 96 animals at a time in multi-well plates, and computer vision software to quantify the animals' locomotor activity, behavioral states, and egg laying events. To demonstrate the capabilities of our system we used it to examine the role of serotonin in C. elegans behavior. We found that egg-laying events are preceded by a period of reduced locomotion, and that this decline in movement requires serotonin signaling. In addition, we identified novel roles of serotonin receptors SER-1 and SER-7 in regulating the effects of serotonin on egg laying across roaming, dwelling, and quiescent locomotor states. Our system will be useful for performing genetic or chemical screens for modulators of behavior.
Collapse
|
18
|
Valdes Michel MF, Phillips BT. SYS-1/beta-catenin inheritance and regulation by Wnt-signaling during asymmetric cell division. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.21.550069. [PMID: 37503055 PMCID: PMC10370182 DOI: 10.1101/2023.07.21.550069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Asymmetric cell division (ACD) allows daughter cells of a polarized mother to acquire different developmental fates. In C. elegans , the Wnt/β-catenin Asymmetry (WβA) pathway oversees many embryonic and larval ACDs; here, a Wnt gradient induces an asymmetric distribution of Wnt signaling components within the dividing mother cell. One terminal nuclear effector of the WβA pathway is the transcriptional activator SYS-1/β-catenin. SYS-1 is sequentially negatively regulated during ACD; first by centrosomal regulation and subsequent proteasomal degradation and second by asymmetric activity of the β-catenin "destruction complex" in one of the two daughter cells, which decreases SYS-1 levels in the absence of WβA signaling. However, the extent to which mother cell SYS-1 influences cell fate decisions of the daughters is unknown. Here, we quantify inherited SYS-1 in the differentiating daughter cells and the role of SYS-1 inheritance in Wnt-directed ACD. Photobleaching experiments demonstrate the GFP::SYS-1 present in daughter cell nuclei is comprised of inherited and de novo translated SYS-1 pools. We used a photoconvertible DENDRA2::SYS-1, to directly observe the dynamics of inherited SYS-1. Photoconversion during mitosis reveals that SYS-1 clearance at the centrosome preferentially degrades older SYS-1, and this accumulation is regulated via dynein trafficking. Photoconversion of the EMS cell during Wnt-driven ACD shows daughter cell inheritance of mother cell SYS-1. Additionally, loss of centrosomal SYS-1 increased inherited SYS-1 and, surprisingly, loss of centrosomal SYS-1 also resulted in increased levels of de novo SYS-1 in both EMS daughter cells. Lastly, we show that daughter cell negative regulation of SYS-1 via the destruction complex member APR-1/APC is key to limit both the de novo and the inherited SYS-1 pools in both the E and the MS cells. We conclude that regulation of both inherited and newly translated SYS-1 via centrosomal processing in the mother cell and daughter cell regulation via Wnt signaling are critical to maintain sister SYS-1 asymmetry during ACD.
Collapse
|
19
|
Gold AL, Hurlock ME, Guevara AM, Isenberg LYZ, Kim Y. Identification of the Polo-like kinase substrate required for homologous synapsis in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607834. [PMID: 39211260 PMCID: PMC11361119 DOI: 10.1101/2024.08.13.607834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The synaptonemal complex (SC) is a zipper-like protein structure that aligns homologous chromosome pairs and regulates recombination during meiosis. Despite its conserved appearance and function, how synapsis occurs between chromosome axes remains elusive. Here, we demonstrate that Polo-like kinases (PLKs) phosphorylate a single conserved residue in the disordered C-terminal tails of two paralogous SC subunits, SYP-5 and SYP-6, to establish an electrostatic interface between the SC central region and chromosome axes in C. elegans . While SYP-5/6 phosphorylation is dispensable for the ability of SC proteins to self-assemble, local phosphorylation by PLKs at the pairing center is crucial for SC elongation between homologous chromosome axes. Additionally, SYP-5/6 phosphorylation is essential for asymmetric SC disassembly and proper PLK-2 localization after crossover designation, which drives chromosome remodeling required for homolog separation during meiosis I. This work identifies a key regulatory mechanism by which localized PLK activity mediates the SC-axis interaction through phosphorylation of SYP-5/6, coupling synapsis initiation to homolog pairing.
Collapse
|
20
|
Hegde S, Modi S, Deihl EW, Glomb OV, Yogev S, Hoerndli FJ, Koushika SP. Axonal mitochondria regulate gentle touch response through control of axonal actin dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607780. [PMID: 39185223 PMCID: PMC11343141 DOI: 10.1101/2024.08.13.607780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Actin in neuronal processes is both stable and dynamic. The origin & functional roles of the different pools of actin is not well understood. We find that mutants that lack mitochondria, ric-7 and mtx-2; miro-1, in neuronal processes also lack dynamic actin. Mitochondria can regulate actin dynamics upto a distance ~80 μm along the neuronal process. Absence of axonal mitochondria and dynamic actin does not markedly alter the Spectrin Membrane Periodic Skeleton (MPS) in touch receptor neurons (TRNs). Restoring mitochondria inTRNs cell autonomously restores dynamic actin in a sod-2 dependent manner. We find that dynamic actin is necessary and sufficient for the localization of gap junction proteins in the TRNs and for the C. elegans gentle touch response. We identify an in vivo mechanism by which axonal mitochondria locally facilitate actin dynamics through reactive oxygen species that we show is necessary for electrical synapses & behaviour.
Collapse
Affiliation(s)
- Sneha Hegde
- Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai-400005, India
| | - Souvik Modi
- Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai-400005, India
| | - Ennis W. Deihl
- Colorado State University, Anatomy and Zoology W309, 1617 Campus Delivery, Fort Collins, 80523 Colorado
| | - Oliver Vinzenz Glomb
- Yale University, Boyer Center for Molecular Medicine, 295 Congress Ave, New Haven, CT 06510
- Current address: Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, 72074 Tübingen, Germany
| | - Shaul Yogev
- Yale University, Boyer Center for Molecular Medicine, 295 Congress Ave, New Haven, CT 06510
| | - Frederic J. Hoerndli
- Colorado State University, Anatomy and Zoology W309, 1617 Campus Delivery, Fort Collins, 80523 Colorado
| | - Sandhya P. Koushika
- Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai-400005, India
| |
Collapse
|
21
|
Jánosi B, Liewald JF, Seidenthal M, Yu SC, Umbach S, Redzovic J, Rentsch D, Alcantara IC, Bergs ACF, Schneider MW, Shao J, Gottschalk A. RIM and RIM-Binding Protein Localize Synaptic CaV2 Channels to Differentially Regulate Transmission in Neuronal Circuits. J Neurosci 2024; 44:e0535222024. [PMID: 38951038 PMCID: PMC11293454 DOI: 10.1523/jneurosci.0535-22.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/16/2024] [Accepted: 05/05/2024] [Indexed: 07/03/2024] Open
Abstract
At chemical synapses, voltage-gated Ca2+ channels (VGCCs) translate electrical signals into a trigger for synaptic vesicle (SV) fusion. VGCCs and the Ca2+ microdomains they elicit must be located precisely to primed SVs to evoke rapid transmitter release. Localization is mediated by Rab3-interacting molecule (RIM) and RIM-binding proteins, which interact and bind to the C terminus of the CaV2 VGCC α-subunit. We studied this machinery at the mixed cholinergic/GABAergic neuromuscular junction of Caenorhabditis elegans hermaphrodites. rimb-1 mutants had mild synaptic defects, through loosening the anchoring of UNC-2/CaV2 and delaying the onset of SV fusion. UNC-10/RIM deletion much more severely affected transmission. Although postsynaptic depolarization was reduced, rimb-1 mutants had increased cholinergic (but reduced GABAergic) transmission, to compensate for the delayed release. This did not occur when the excitation-inhibition (E-I) balance was altered by removing GABA transmission. Further analyses of GABA defective mutants and GABAA or GABAB receptor deletions, as well as cholinergic rescue of RIMB-1, emphasized that GABA neurons may be more affected than cholinergic neurons. Thus, RIMB-1 function differentially affects excitation-inhibition balance in the different motor neurons, and RIMB-1 thus may differentially regulate transmission within circuits. Untethering the UNC-2/CaV2 channel by removing its C-terminal PDZ ligand exacerbated the rimb-1 defects, and similar phenotypes resulted from acute degradation of the CaV2 β-subunit CCB-1. Therefore, untethering of the CaV2 complex is as severe as its elimination, yet it does not abolish transmission, likely due to compensation by CaV1. Thus, robustness and flexibility of synaptic transmission emerge from VGCC regulation.
Collapse
Affiliation(s)
- Barbara Jánosi
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Jana F Liewald
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Marius Seidenthal
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Szi-Chieh Yu
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Simon Umbach
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Jasmina Redzovic
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Dennis Rentsch
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Ivan C Alcantara
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Amelie C F Bergs
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Martin W Schneider
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Jiajie Shao
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| |
Collapse
|
22
|
Chen S, Phillips CM. Nuclear Argonaute protein NRDE-3 switches small RNA binding partners during embryogenesis coincident with the formation of SIMR granules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605686. [PMID: 39131395 PMCID: PMC11312606 DOI: 10.1101/2024.07.29.605686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
RNA interference (RNAi) is a conserved gene regulation mechanism that utilizes the Argonaute protein and their associated small RNAs to exert regulatory function on complementary transcripts. While the majority of germline-expressed RNAi pathway components reside in perinuclear germ granules, it is unknown whether and how RNAi pathways are spatially organized in other cell types. Here we find that the small RNA biogenesis machinery is spatially and temporally organized during embryogenesis. Specifically, the RNAi factor, SIMR-1, forms visible concentrates during mid-embryogenesis that contain an RNA-dependent RNA polymerase, a poly-UG polymerase, and the unloaded nuclear Argonaute protein, NRDE-3. Further, we observe that many other RNAi factors form foci in embryonic cells distinct from SIMR granules, including the Argonaute protein CSR-1, underscoring a potential role for cytoplasmic concentrates of RNAi factors to promote gene regulation in embryos. Curiously, coincident with the appearance of the "SIMR granules", the small RNAs bound to NRDE-3 switch from predominantly CSR-class 22G-RNAs to ERGO-dependent 22G-RNAs. Thus, our study defines two separable roles for NRDE-3, targeting germline-expressed genes during early embryogenesis and switching later in embryogenesis to repress recently duplicated genes and retrotransposons in somatic cells, highlighting the plasticity of Argonaute proteins and the need for more precise temporal characterization of Argonaute-small RNA interactions.
Collapse
Affiliation(s)
- Shihui Chen
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089
| | - Carolyn M Phillips
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
23
|
Kim AJ, Miller SI, Greiner EC, Kettenbach AN, Griffin EE. PLK-1 regulates MEX-1 polarization in the C. elegans zygote. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605193. [PMID: 39091813 PMCID: PMC11291152 DOI: 10.1101/2024.07.26.605193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The one-cell C. elegans embryo undergoes an asymmetric cell division during which germline factors such as the RNA-binding proteins POS-1 and MEX-1 segregate to the posterior cytoplasm, leading to their asymmetric inheritance to the posterior germline daughter cell. Previous studies found that the RNA-binding protein MEX-5 recruits polo-like kinase PLK-1 to the anterior cytoplasm where PLK-1 inhibits the retention of its substrate POS-1, leading to POS-1 segregation to the posterior. In this study, we tested whether PLK-1 similarly regulates MEX-1 polarization. We find that both the retention of MEX-1 in the anterior and the segregation of MEX-1 to the posterior depend on PLK kinase activity and on the interaction between MEX-5 and PLK-1. Human PLK1 directly phosphorylates recombinant MEX-1 on 9 predicted PLK-1 sites in vitro, four of which were identified in previous phosphoproteomic analysis of C. elegans embryos. The introduction of alanine substitutions at these four PLK-1 phosphorylation sites (MEX-1(4A)) significantly weakened the inhibition of MEX-1 retention in the anterior, thereby weakening MEX-1 segregation to the posterior. In contrast, mutation of a predicted CDK1 phosphorylation site had no effect on MEX-1 retention or on MEX-1 segregation. MEX-1(4A) mutants are viable and fertile but display significant sterility and fecundity defects at elevated temperatures. Taken together with our previous findings, these findings suggest PLK-1 phosphorylation drives both MEX-1 and POS-1 polarization during the asymmetric division of the zygote.
Collapse
Affiliation(s)
- Amelia J. Kim
- Department of Biological Sciences, Dartmouth College, Hanover NH 03755
| | | | - Elora C. Greiner
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover NH 03755
- Dartmouth Cancer Center, Lebanon NH 03755
| | - Arminja N. Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover NH 03755
- Dartmouth Cancer Center, Lebanon NH 03755
| | - Erik E. Griffin
- Department of Biological Sciences, Dartmouth College, Hanover NH 03755
- School of Life Sciences, University of Warwick, Coventry UK
| |
Collapse
|
24
|
Aguilar GR, Vidal B, Ji H, Evenblij J, Ji H, Valperga G, Liao CP, Fang-Yen C, Hobert O. Functional analysis of conserved C. elegans bHLH family members uncovers lifespan control by a peptidergic hub neuron. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603289. [PMID: 39071424 PMCID: PMC11275782 DOI: 10.1101/2024.07.12.603289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Throughout the animal kingdom, several members of the basic helix-loop-helix (bHLH) family act as proneural genes during early steps of nervous system development. Roles of bHLH genes in specifying terminal differentiation of postmitotic neurons have been less extensively studied. We analyze here the function of five C. elegans bHLH genes, falling into three phylogenetically conserved subfamilies, which are continuously expressed in a very small number of postmitotic neurons in the central nervous system. We show (a) that two orthologs of the vertebrate bHLHb4/b5 genes, called hlh-17 and hlh-32, function redundantly to specify the identity of a single head interneuron (AUA), as well as an individual motor neuron (VB2), (b) that the PTF1a ortholog hlh-13 acts as a terminal selector to control terminal differentiation and function of the sole octopaminergic neuron class in C. elegans, RIC, and (c) that the NHLH1/2 ortholog hlh-15 controls terminal differentiation and function of the peptidergic AVK head interneuron class, a known neuropeptidergic signaling hub in the animal. Strikingly, through null mutant analysis and cell-specific rescue experiments, we find that loss of hlh-15/NHLH in the peptidergic AVK neurons and the resulting abrogation of neuropeptide secretion causes a substantially expanded lifespan of the animal, revealing an unanticipated impact of a central, peptidergic hub neuron in regulating lifespan, which we propose to be akin to hypothalamic control of lifespan in vertebrates. Taken together, our functional analysis reveals themes of bHLH gene function during terminal differentiation that are complementary to the earlier lineage specification roles of other bHLH family members. However, such late functions are much more sparsely employed by members of the bHLH transcription factor family, compared to the function of the much more broadly employed homeodomain transcription factor family.
Collapse
Affiliation(s)
- G. Robert Aguilar
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| | - Berta Vidal
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| | - Hongzhu Ji
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| | - Joke Evenblij
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
- Technische Universität, Braunschweig, Germany
| | - Hongfei Ji
- Department of Biomedical Engineering, Ohio State University, Columbus, OH
| | - Giulio Valperga
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| | - Chien-Po Liao
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| | | | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| |
Collapse
|
25
|
Palominos MF, Bharadwaj R, Tralka C, Trang K, Aka D, Alami M, Andrews D, Bartlett BI, Golde C, Liu J, Le-Pedroza M, Perrot R, Seiter B, Sparrow C, Shapira M, Martin CH. The West African lungfish secretes a living cocoon during aestivation with uncertain antimicrobial function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.602297. [PMID: 39026789 PMCID: PMC11257426 DOI: 10.1101/2024.07.05.602297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
One of the most exceptional adaptations to extreme drought is found in the sister group to tetrapods, the lungfishes (Dipnoi), which can aestivate inside a mucus cocoon for multiple years at reduced metabolic rates with complete cessation of ingestion and excretion. However, the function of the cocoon tissue is not fully understood. Here we developed a new more natural laboratory protocol for inducing aestivation in the West African lungfish, Protopterus annectens, and investigated the structure and function of the cocoon. We used electron microscopy and imaging of live tissue-stains to confirm that the inner and outer layers of the paper-thin cocoon are composed primarily of living cells. However, we also repeatedly observed extensive bacterial and fungal growth covering the cocoon and found no evidence of anti-microbial activity in vitro against E. coli for the cocoon tissue in this species. This classroom discovery-based research, performed during a course-based undergraduate research experience course (CURE), provides a robust laboratory protocol for investigating aestivation and calls into the question the function of this bizarre vertebrate adaptation.
Collapse
Affiliation(s)
- M Fernanda Palominos
- Department of Integrative Biology, University of California, Berkeley, CA 94720
- Museum of Vertebrate Zoology, University of California, Berkeley, CA 94720
| | | | - Charles Tralka
- Department of Integrative Biology, University of California, Berkeley, CA 94720
- Museum of Vertebrate Zoology, University of California, Berkeley, CA 94720
| | - Kenneth Trang
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - David Aka
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Mariam Alami
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Dominique Andrews
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Ben I Bartlett
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Chloe Golde
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Joseph Liu
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Maya Le-Pedroza
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Robert Perrot
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Blanca Seiter
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Claudia Sparrow
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Michael Shapira
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Christopher H Martin
- Department of Integrative Biology, University of California, Berkeley, CA 94720
- Museum of Vertebrate Zoology, University of California, Berkeley, CA 94720
| |
Collapse
|
26
|
Kong JN, Dipon Ghosh D, Savvidis A, Sando SR, Droste R, Robert Horvitz H. Transcriptional landscape of a hypoxia response identifies cell-specific pathways for adaptation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601765. [PMID: 39005398 PMCID: PMC11245032 DOI: 10.1101/2024.07.02.601765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
How the HIF-1 (Hypoxia-Inducible) transcription factor drives and coordinates distinct responses to low oxygen across diverse cell types is poorly understood. We present a multi-tissue single-cell gene-expression atlas of the hypoxia response of the nematode Caenorhabditis elegans . This atlas highlights how cell-type-specific HIF-1 responses overlap and diverge among and within neuronal, intestinal, and muscle tissues. Using the atlas to guide functional analyses of candidate muscle-specific HIF-1 effectors, we discovered that HIF-1 activation drives downregulation of the tspo-1 ( TSPO, Translocator Protein) gene in vulval muscle cells to modulate a hypoxia-driven change in locomotion caused by contraction of body-wall muscle cells. We further showed that in human cardiomyocytes HIF-1 activation decreases levels of TSPO and thereby alters intracellular cholesterol transport and the mitochondrial network. We suggest that TSPO-1 is an evolutionarily conserved mediator of HIF-1-dependent modulation of muscle and conclude that our gene-expression atlas can help reveal how HIF-1 drives cell-specific adaptations to hypoxia.
Collapse
|
27
|
Elaswad MT, Gao M, Tice VE, Bright CG, Thomas GM, Munderloh C, Trombley NJ, Haddad CN, Johnson UG, Cichon AN, Schisa JA. The CCT chaperonin and actin modulate the ER and RNA-binding protein condensation during oogenesis to maintain translational repression of maternal mRNA and oocyte quality. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601596. [PMID: 39005301 PMCID: PMC11244991 DOI: 10.1101/2024.07.01.601596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The regulation of maternal mRNAs is essential for proper oogenesis, the production of viable gametes, and to avoid birth defects and infertility. Many oogenic RNA-binding proteins have been identified with roles in mRNA metabolism, some of which localize to dynamic ribonucleoprotein granules and others that appear dispersed. Here, we use a combination of in vitro condensation assays and the in vivo C. elegans oogenesis model to determine the intrinsic properties of the conserved KH-domain MEX-3 protein and to identify novel regulators of MEX-3 and the Lsm protein, CAR-1. We demonstrate that MEX-3 undergoes liquid-liquid phase separation and appears to have intrinsic gel-like properties in vitro . We also identify novel roles for the CCT chaperonin and actin in preventing ectopic RNA-binding protein condensates in maturing oocytes that appear to be independent of MEX-3 folding. CCT and actin also oppose the expansion of ER sheets that may promote ectopic condensation of RNA-binding proteins that are associated with de-repression of maternal mRNA. This regulatory network is essential to preserve oocyte quality, prevent infertility, and may have implications for understanding the role of hMex3 phase transitions in cancer. Significance statement The molecular mechanisms that regulate phase transitions of oogenic RNA-binding proteins are critical to elucidate but are not fully understood.We identify novel regulators of RNA-binding protein phase transitions in maturing oocytes that are required to maintain translational repression of maternal mRNAs and oocyte quality.This study is the first to elucidate a regulatory network involving the CCT chaperonin, actin, and the ER for phase transitions of RNA-binding proteins during oogenesis. Our findings for the conserved MEX-3 protein may also be applicable to better understanding the role of hMex3 phase transitions in cancer.
Collapse
|
28
|
Baccas M, Ganesan V, Leung A, Pineiro L, McKillop AN, Liu J. SEM-2/SoxC regulates multiple aspects of C. elegans postembryonic mesoderm development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.04.602042. [PMID: 39005444 PMCID: PMC11245110 DOI: 10.1101/2024.07.04.602042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Development of multicellular organisms requires well-orchestrated interplay between cell-intrinsic transcription factors and cell-cell signaling. One set of highly conserved transcription factors that plays diverse roles in development is the SoxC group. C. elegans contains a sole SoxC protein, SEM-2. SEM-2 is essential for embryonic development, and for specifying the sex myoblast (SM) fate in the postembryonic mesoderm, the M lineage. We have identified a novel partial loss-of-function sem-2 allele that has a proline to serine change in the C-terminal tail of the highly conserved DNA-binding domain. Detailed analyses of mutant animals harboring this point mutation uncovered new functions of SEM-2 in the M lineage. First, SEM-2 functions antagonistically with LET-381, the sole C. elegans FoxF/C forkhead transcription factor, to regulate dorsoventral patterning of the M lineage. Second, in addition to specifying the SM fate, SEM-2 is essential for the proliferation and diversification of the SM lineage. Finally, SEM-2 appears to directly regulate the expression of hlh-8, which encodes a basic helix-loop-helix Twist transcription factor and plays critical roles in proper patterning of the M lineage. Our data, along with previous studies, suggest an evolutionarily conserved relationship between SoxC and Twist proteins. Furthermore, our work identified new interactions in the gene regulatory network (GRN) underlying C. elegans postembryonic development and adds to the general understanding of the structure-function relationship of SoxC proteins.
Collapse
Affiliation(s)
- Marissa Baccas
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Vanathi Ganesan
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Amy Leung
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Lucas Pineiro
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | | | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| |
Collapse
|
29
|
Connors CQ, Martin SL, Dumont J, Shirasu-Hiza M, Canman JC. Cell type-specific regulation by different cytokinetic pathways in the early embryo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601054. [PMID: 38979134 PMCID: PMC11230459 DOI: 10.1101/2024.06.27.601054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Cytokinesis, the physical division of one cell into two, is typically assumed to use the same molecular process across animal cells. However, regulation of cell division can vary significantly among different cell types, even within the same multicellular organism. Using six fast-acting temperature-sensitive (ts) cytokinesis-defective mutants, we found that each had unique cell type-specific profiles in the early C. elegans embryo. Certain cell types were more sensitive than others to actomyosin and spindle signaling disruptions, disrupting two members of the same complex could result in different phenotypes, and protection against actomyosin inhibition did not always protect against spindle signaling inhibition.
Collapse
|
30
|
Ji H, Chen D, Fang-Yen C. Segmentation-free measurement of locomotor frequency in Caenorhabditis elegans using image invariants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.16.575892. [PMID: 38293059 PMCID: PMC10827210 DOI: 10.1101/2024.01.16.575892] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
An animal's locomotor rate is an important indicator of its motility. In studies of the nematode C. elegans, assays of the frequency of body bending waves have often been used to discern the effects of mutations, drugs, or aging. Traditional manual methods for measuring locomotor frequency are low in throughput and subject to human error. Most current automated methods depend on image segmentation, which requires high image quality and is prone to errors. Here, we describe an algorithm for automated estimation of C. elegans locomotor frequency using image invariants, i.e., shape-based parameters that are independent of object translation, rotation, and scaling. For each video frame, the method calculates a combination of 8 Hu's moment invariants and a set of Maximally Stable Extremal Regions (MSER) invariants. The algorithm then calculates the locomotor frequency by computing the autocorrelation of the time sequence of the invariant ensemble. Results of our method show excellent agreement with manual or segmentation-based results over a wide range of frequencies. We show that compared to a segmentation-based method that analyzes a worm's shape and a method based on video covariance, our technique is more robust to low image quality and background noise. We demonstrate the system's capabilities by testing the effects of serotonin and serotonin pathway mutations on C. elegans locomotor frequency.
Collapse
|
31
|
Kolli S, Kline CJ, Rad KM, Wehman AM. Phagolysosomes break down the membrane of a non-apoptotic corpse independent of macroautophagy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599770. [PMID: 38948720 PMCID: PMC11212964 DOI: 10.1101/2024.06.19.599770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Cell corpses must be cleared in an efficient manner to maintain tissue homeostasis and regulate immune responses. Ubiquitin-like Atg8/LC3 family proteins promote the degradation of membranes and internal cargo during both macroautophagy and corpse clearance, raising the question how macroautophagy contributes to corpse clearance. Studying the clearance of non-apoptotic dying polar bodies in Caenorhabditis elegans embryos, we show that the LC3 ortholog LGG-2 is enriched in the polar body phagolysosome independent of membrane association or autophagosome formation. We demonstrate that ATG-16.1 and ATG-16.2, which promote membrane association of lipidated Atg8/LC3 proteins, redundantly promote polar body membrane breakdown in phagolysosomes independent of their role in macroautophagy. We also show that the lipid scramblase ATG-9 is needed for autophagosome formation in early embryos but is dispensable for timely polar body membrane breakdown or protein cargo degradation. These findings demonstrate that macroautophagy is not required to promote polar body degradation, in contrast to recent findings with apoptotic corpse clearance in C. elegans embryos. Determining how membrane association of Atg8/LC3 promotes the breakdown of different types of cell corpses in distinct cell types or metabolic states is likely to give insights into the mechanisms of immunoregulation during normal development, physiology, and disease.
Collapse
Affiliation(s)
- Shruti Kolli
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Cassidy J. Kline
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Kimya M. Rad
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Ann M. Wehman
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| |
Collapse
|
32
|
Packer J, Gubieda AG, Brooks A, Deutz LN, Squires I, Ellison S, Schneider C, Naganathan SR, Wollman AJ, Dickinson DJ, Rodriguez J. Atypical Protein Kinase C Promotes its own Asymmetric Localisation by Phosphorylating Cdc42 in the C. elegans zygote. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.27.563985. [PMID: 38009101 PMCID: PMC10675845 DOI: 10.1101/2023.10.27.563985] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
Atypical protein kinase C (aPKC) is a major regulator of cell polarity. Acting in conjunction with Par6, Par3 and the small GTPase Cdc42, aPKC becomes asymmetrically localised and drives the polarisation of cells. aPKC activity is crucial for its own asymmetric localisation, suggesting a hitherto unknown feedback mechanism contributing to polarisation. Here we show in the C. elegans zygote that the feedback relies on aPKC phosphorylation of Cdc42 at serine 71. The turnover of CDC-42 phosphorylation ensures optimal aPKC asymmetry and activity throughout polarisation by tuning Par6/aPKC association with Par3 and Cdc42. Moreover, turnover of Cdc42 phosphorylation regulates actomyosin cortex dynamics that are known to drive aPKC asymmetry. Given the widespread role of aPKC and Cdc42 in cell polarity, this form of self-regulation of aPKC may be vital for the robust control of polarisation in many cell types.
Collapse
Affiliation(s)
- John Packer
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- These authors contributed equally
| | - Alicia G. Gubieda
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- These authors contributed equally
| | - Aaron Brooks
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- These authors contributed equally
| | - Lars N. Deutz
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
- These authors contributed equally
| | - Iolo Squires
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- These authors contributed equally
| | | | | | - Sundar Ram Naganathan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Adam J.M. Wollman
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Daniel J. Dickinson
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Josana Rodriguez
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Lead contact
| |
Collapse
|
33
|
Qiu C, Crittenden SL, Carrick BH, Dillard LB, Costa Dos Santos SJ, Dandey VP, Dutcher RC, Viverette EG, Wine RN, Woodworth J, Campbell ZT, Wickens M, Borgnia MJ, Kimble J, Tanaka Hall TM. A higher order PUF complex is central to regulation of C. elegans germline stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.599074. [PMID: 38915480 PMCID: PMC11195197 DOI: 10.1101/2024.06.14.599074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
PUF RNA-binding proteins are broadly conserved stem cell regulators. Nematode PUF proteins maintain germline stem cells (GSCs) and, with key partner proteins, repress differentiation mRNAs, including gld-1. Here we report that PUF protein FBF-2 and its partner LST-1 form a ternary complex that represses gld-1 via a pair of adjacent FBF-2 binding elements (FBEs) in its 3ÚTR. One LST-1 molecule links two FBF-2 molecules via motifs in the LST-1 intrinsically-disordered region; the gld-1 FBE pair includes a well-established 'canonical' FBE and a newly-identified noncanonical FBE. Remarkably, this FBE pair drives both full RNA repression in GSCs and full RNA activation upon differentiation. Discovery of the LST-1-FBF-2 ternary complex, the gld-1 adjacent FBEs, and their in vivo significance predicts an expanded regulatory repertoire of different assemblies of PUF-partner complexes in nematode germline stem cells. It also suggests analogous PUF controls may await discovery in other biological contexts and organisms.
Collapse
Affiliation(s)
- Chen Qiu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | | | - Brian H. Carrick
- Department of Biochemistry, University of Wisconsin, Madison, WI 53706, USA
- Current address: MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Lucas B. Dillard
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
- Current address: Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephany J. Costa Dos Santos
- Department of Biochemistry, University of Wisconsin, Madison, WI 53706, USA
- These authors contributed equally to the manuscript and are listed in alphabetical order
| | - Venkata P. Dandey
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
- These authors contributed equally to the manuscript and are listed in alphabetical order
| | - Robert C. Dutcher
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
- These authors contributed equally to the manuscript and are listed in alphabetical order
| | - Elizabeth G. Viverette
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
- These authors contributed equally to the manuscript and are listed in alphabetical order
- Current address: Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert N. Wine
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
- These authors contributed equally to the manuscript and are listed in alphabetical order
| | - Jennifer Woodworth
- Department of Biochemistry, University of Wisconsin, Madison, WI 53706, USA
- These authors contributed equally to the manuscript and are listed in alphabetical order
| | - Zachary T. Campbell
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Marvin Wickens
- Department of Biochemistry, University of Wisconsin, Madison, WI 53706, USA
| | - Mario J. Borgnia
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Judith Kimble
- Department of Biochemistry, University of Wisconsin, Madison, WI 53706, USA
| | - Traci M. Tanaka Hall
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
- Lead contact
| |
Collapse
|
34
|
Caro L, Wei AD, Thomas CA, Posch G, Uremis A, Franzi MC, Abell SJ, Laszlo AH, Gundlach JH, Ramirez JM, Ailion M. Mechanism of an animal toxin-antidote system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598564. [PMID: 38915716 PMCID: PMC11195288 DOI: 10.1101/2024.06.11.598564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Toxin-antidote systems are selfish genetic elements composed of a linked toxin and antidote. The peel-1 zeel-1 toxin-antidote system in C. elegans consists of a transmembrane toxin protein PEEL-1 which acts cell autonomously to kill cells. Here we investigate the molecular mechanism of PEEL-1 toxicity. We find that PEEL-1 requires a small membrane protein, PMPL-1, for toxicity. Together, PEEL-1 and PMPL-1 are sufficient for toxicity in a heterologous system, HEK293T cells, and cause cell swelling and increased cell permeability to monovalent cations. Using purified proteins, we show that PEEL-1 and PMPL-1 allow ion flux through lipid bilayers and generate currents which resemble ion channel gating. Our work suggests that PEEL-1 kills cells by co-opting PMPL-1 and creating a cation channel.
Collapse
Affiliation(s)
- Lews Caro
- Molecular and Cellular Biology Ph.D. Program, University of Washington; Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington; Seattle, WA 91895, USA
| | - Aguan D. Wei
- Norcliffe Foundation Center for Integrative Brain Research, Seattle Children’s Research Institute; Seattle, WA 98101, USA
| | | | - Galen Posch
- Department of Biochemistry, University of Washington; Seattle, WA 91895, USA
| | - Ahmet Uremis
- Department of Biochemistry, University of Washington; Seattle, WA 91895, USA
| | | | - Sarah J. Abell
- Department of Physics, University of Washington; Seattle, WA 91895, USA
| | - Andrew H. Laszlo
- Department of Physics, University of Washington; Seattle, WA 91895, USA
| | - Jens H. Gundlach
- Department of Physics, University of Washington; Seattle, WA 91895, USA
| | - Jan-Marino Ramirez
- Norcliffe Foundation Center for Integrative Brain Research, Seattle Children’s Research Institute; Seattle, WA 98101, USA
- Department of Neurological Surgery, University of Washington School of Medicine; Seattle, WA 98104, USA
| | - Michael Ailion
- Molecular and Cellular Biology Ph.D. Program, University of Washington; Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington; Seattle, WA 91895, USA
| |
Collapse
|
35
|
Riordan R, Saxton A, McMillan PJ, Kow RL, Liachko NF, Kraemer BC. TMEM106B C-terminal fragments aggregate and drive neurodegenerative proteinopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598478. [PMID: 38915598 PMCID: PMC11195232 DOI: 10.1101/2024.06.11.598478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Genetic variation in the lysosomal and transmembrane protein 106B (TMEM106B) modifies risk for a diverse range of neurodegenerative disorders, especially frontotemporal lobar degeneration (FTLD) with progranulin (PGRN) haplo-insufficiency, although the molecular mechanisms involved are not yet understood. Through advances in cryo-electron microscopy (cryo-EM), homotypic aggregates of the C-Terminal domain of TMEM106B (TMEM CT) were discovered as a previously unidentified cytosolic proteinopathy in the brains of FTLD, Alzheimer's disease, progressive supranuclear palsy (PSP), and dementia with Lewy bodies (DLB) patients. While it remains unknown what role TMEM CT aggregation plays in neuronal loss, its presence across a range of aging related dementia disorders indicates involvement in multi-proteinopathy driven neurodegeneration. To determine the TMEM CT aggregation propensity and neurodegenerative potential, we characterized a novel transgenic C. elegans model expressing the human TMEM CT fragment constituting the fibrillar core seen in FTLD cases. We found that pan-neuronal expression of human TMEM CT in C. elegans causes neuronal dysfunction as evidenced by behavioral analysis. Cytosolic aggregation of TMEM CT proteins accompanied the behavioral dysfunction driving neurodegeneration, as illustrated by loss of GABAergic neurons. To investigate the molecular mechanisms driving TMEM106B proteinopathy, we explored the impact of PGRN loss on the neurodegenerative effect of TMEM CT expression. To this end, we generated TMEM CT expressing C. elegans with loss of pgrn-1, the C. elegans ortholog of human PGRN. Neither full nor partial loss of pgrn-1 altered the motor phenotype of our TMEM CT model suggesting TMEM CT aggregation occurs downstream of PGRN loss of function. We also tested the ability of genetic suppressors of tauopathy to rescue TMEM CT pathology. We found that genetic knockout of spop-1, sut-2, and sut-6 resulted in weak to no rescue of proteinopathy phenotypes, indicating that the mechanistic drivers of TMEM106B proteinopathy may be distinct from tauopathy. Taken together, our data demonstrate that TMEM CT aggregation can kill neurons. Further, expression of TMEM CT in C. elegans neurons provides a useful model for the functional characterization of TMEM106B proteinopathy in neurodegenerative disease.
Collapse
Affiliation(s)
- Ruben Riordan
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Aleen Saxton
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Pamela J. McMillan
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington 98195, USA
| | - Rebecca L Kow
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Nicole F. Liachko
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Brian C. Kraemer
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington 98195, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
36
|
Clites BL, Frohock B, Koury EJ, Andersen EC, Pierce JT. Natural variation in protein kinase D modifies alcohol sensitivity in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598102. [PMID: 38895441 PMCID: PMC11185769 DOI: 10.1101/2024.06.09.598102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Differences in naïve alcohol sensitivity between individuals are a strong predictor of later life alcohol use disorders (AUD). However, the genetic bases for alcohol sensitivity (beyond ethanol metabolism) and pharmacological approaches to modulate alcohol sensitivity remain poorly understood. We used a high-throughput behavioral screen to measure acute behavioral sensitivity to alcohol, a model of intoxication, in a genetically diverse set of over 150 wild strains of the nematode Caenorhabditis elegans. We performed a genome-wide association study to identify loci that underlie natural variation in alcohol sensitivity. We identified five quantitative trait loci (QTL) and further show that variants in the C. elegans ortholog of protein kinase D, dkf-2, likely underlie the chromosome V QTL. We found that resistance to intoxication was conferred by dkf-2 loss-of-function mutations as well as partly by a PKD inhibitor in a dkf-2-dependent manner. Protein kinase D might represent a conserved, druggable target to modify alcohol sensitivity with application towards AUD.
Collapse
Affiliation(s)
- Benjamin L Clites
- Waggoner Center for Alcohol & Addiction Research, Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin TX
| | - Brooke Frohock
- Waggoner Center for Alcohol & Addiction Research, Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin TX
| | - Emily J Koury
- Department of Biology, Johns Hopkins University, Baltimore MD
| | - Erik C Andersen
- Department of Biology, Johns Hopkins University, Baltimore MD
| | - Jonathan T Pierce
- Waggoner Center for Alcohol & Addiction Research, Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin TX
| |
Collapse
|
37
|
Knudsen-Palmer DR, Raman P, Ettefa F, De Ravin L, Jose AM. Target-specific requirements for RNA interference can arise through restricted RNA amplification despite the lack of specialized pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.07.527351. [PMID: 36798330 PMCID: PMC9934570 DOI: 10.1101/2023.02.07.527351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Since double-stranded RNA (dsRNA) is effective for silencing a wide variety of genes, all genes are typically considered equivalent targets for such RNA interference (RNAi). Yet, loss of some regulators of RNAi in the nematode C. elegans can selectively impair the silencing of some genes. Here we show that such selective requirements can be explained by an intersecting network of regulators acting on genes with differences in their RNA metabolism. In this network, the Maelstrom domain-containing protein RDE-10, the intrinsically disordered protein MUT-16, and the Argonaute protein NRDE-3 work together so that any two are required for silencing one somatic gene, but each is singly required for silencing another somatic gene, where only the requirement for NRDE-3 can be overcome by enhanced dsRNA processing. Quantitative models and their exploratory simulations led us to find that (1) changing cis-regulatory elements of the target gene can reduce the dependence on NRDE-3, (2) animals can recover from silencing in non-dividing cells and (3) cleavage and tailing of mRNAs with UG dinucleotides, which makes them templates for amplifying small RNAs, is enriched within 'pUG zones' matching the dsRNA. Similar crosstalk between pathways and restricted amplification could result in apparently selective silencing by endogenous RNAs.
Collapse
Affiliation(s)
- Daphne R. Knudsen-Palmer
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, USA. Biological Sciences Graduate Program, University of Maryland, College Park, USA
| | - Pravrutha Raman
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, USA. Biological Sciences Graduate Program, University of Maryland, College Park, USA
- Current address: Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Farida Ettefa
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, USA. Biological Sciences Graduate Program, University of Maryland, College Park, USA
- Current address: Institute for Systems Genetics, New York University School of Medicine, New York, NY, USA
| | - Laura De Ravin
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, USA. Biological Sciences Graduate Program, University of Maryland, College Park, USA
| | - Antony M. Jose
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, USA. Biological Sciences Graduate Program, University of Maryland, College Park, USA
| |
Collapse
|
38
|
Leuthner TC, Zhang S, Kohrn BF, Stapleton HM, Baugh LR. Structure-specific variation in per- and polyfluoroalkyl substances toxicity among genetically diverse Caenorhabditis elegans strains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596269. [PMID: 38854041 PMCID: PMC11160736 DOI: 10.1101/2024.05.29.596269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background There are >14,500 structurally diverse per- and polyfluoroalkyl substances (PFAS). Despite knowledge that these "forever chemicals" are in 99% of humans, mechanisms of toxicity and adverse health effects are incompletely known. Furthermore, the contribution of genetic variation to PFAS susceptibility and health consequences is unknown. Objectives We determined the toxicity of a structurally distinct set of PFAS in twelve genetically diverse strains of the genetic model system Caenorhabditis elegans. Methods Dose-response curves for four perfluoroalkyl carboxylic acids (PFNA, PFOA, PFPeA, and PFBA), two perfluoroalkyl sulfonic acids (PFOS and PFBS), two perfluoroalkyl sulfonamides (PFOSA and PFBSA), two fluoroether carboxylic acids (GenX and PFMOAA), one fluoroether sulfonic acid (PFEESA), and two fluorotelomers (6:2 FCA and 6:2 FTS) were determined in the C. elegans laboratory reference strain, N2, and eleven genetically diverse wild strains. Body length was quantified by image analysis at each dose after 48 hr of developmental exposure of L1 arrest-synchronized larvae to estimate effective concentration values (EC50). Results There was a significant range in toxicity among PFAS: PFOSA > PFBSA ≈ PFOS ≈ PFNA > PFOA > GenX ≈ PFEESA > PFBS ≈ PFPeA ≈ PFBA. Long-chain PFAS had greater toxicity than short-chain, and fluorosulfonamides were more toxic than carboxylic and sulfonic acids. Genetic variation explained variation in susceptibility to PFBSA, PFOS, PFBA, PFOA, GenX, PFEESA, PFPeA, and PFBA. There was significant variation in toxicity among C. elegans strains due to chain length, functional group, and between legacy and emerging PFAS. Conclusion C. elegans respond to legacy and emerging PFAS of diverse structures, and this depends on specific structures and genetic variation. Harnessing the natural genetic diversity of C. elegans and the structural complexity of PFAS is a powerful New Approach Methodology (NAM) to investigate structure-activity relationships and mechanisms of toxicity which may inform regulation of other PFAS to improve human and environmental health.
Collapse
Affiliation(s)
- Tess C. Leuthner
- Department of Biology, Duke University, Durham, North Carolina, USA
| | - Sharon Zhang
- Nicholas School of the Environment, Duke University, Durham, North Carolina, USA
| | - Brendan F Kohrn
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Heather M. Stapleton
- Nicholas School of the Environment, Duke University, Durham, North Carolina, USA
| | - L. Ryan Baugh
- Department of Biology, Duke University, Durham, North Carolina, USA
- Center for Genomic and Computational Biology, Duke University, North Carolina, USA
| |
Collapse
|
39
|
Catela C, Assimacopoulos S, Chen Y, Tsioras K, Feng W, Kratsios P. The Iroquois ( Iro/Irx) homeobox genes are conserved Hox targets involved in motor neuron development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596714. [PMID: 38853975 PMCID: PMC11160718 DOI: 10.1101/2024.05.30.596714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The Iroquois (Iro/Irx) homeobox genes encode transcription factors with fundamental roles in animal development. Despite their link to various congenital conditions in humans, our understanding of Iro/Irx gene expression, function, and regulation remains incomplete. Here, we conducted a systematic expression analysis of all six mouse Irx genes in the embryonic spinal cord. We found five Irx genes (Irx1, Irx2, Irx3, Irx5, and Irx6) to be confined mostly to ventral spinal domains, offering new molecular markers for specific groups of post-mitotic motor neurons (MNs). Further, we engineered Irx2, Irx5, and Irx6 mouse mutants and uncovered essential but distinct roles for Irx2 and Irx6 in MN development. Last, we found that the highly conserved regulators of MN development across species, the HOX proteins, directly control Irx gene expression both in mouse and C. elegans MNs, critically expanding the repertoire of HOX target genes in the developing nervous system. Altogether, our study provides important insights into Iro/Irx expression and function in the developing spinal cord, and uncovers an ancient gene regulatory relationship between HOX and Iro/Irx genes.
Collapse
Affiliation(s)
- Catarina Catela
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
- Neuroscience Institute, University of Chicago, Chicago, IL, USA
| | - Stavroula Assimacopoulos
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
- Neuroscience Institute, University of Chicago, Chicago, IL, USA
| | - Yihan Chen
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
- Neuroscience Institute, University of Chicago, Chicago, IL, USA
| | - Konstantinos Tsioras
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
- Neuroscience Institute, University of Chicago, Chicago, IL, USA
| | - Weidong Feng
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
- Neuroscience Institute, University of Chicago, Chicago, IL, USA
| | - Paschalis Kratsios
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
- Neuroscience Institute, University of Chicago, Chicago, IL, USA
| |
Collapse
|
40
|
Gajjar G, Huggins HP, Kim ES, Huang W, Bonnet FX, Updike DL, Keiper BD. Two germ granule eIF4E isoforms reside in different mRNPs to hand off C elegans mRNAs from translational repression to activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595216. [PMID: 38826235 PMCID: PMC11142241 DOI: 10.1101/2024.05.24.595216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
We studied the function of translation factor eIF4E isoforms in regulating mRNAs in germ cell granules/condensates. Translational control of mRNAs plays an essential role in germ cell gene regulation. Messenger ribonucleoprotein (mRNP) complexes assemble on mRNAs as they move from the nucleus into perinuclear germ granules to exert both positive and negative post-transcriptional regulation in the cytoplasm. In C. elegans , germ granules are surprisingly dynamic mRNP condensates that remodel during development. Two eIF4E isoforms (called IFE-1 and IFE-3), eIF4E-Interacting Proteins (4EIPs), RBPs, DEAD-box helicases, polyadenylated mRNAs, Argonautes and miRNAs all occupy positions in germ granules. Affinity purification of IFE-1 and IFE-3 allowed mass spectrometry and mRNA-Seq to identify the proteins and mRNAs that populate stable eIF4E mRNPs. We find translationally controlled mRNAs (e.g. pos-1, mex-3, spn-4, etc.) enriched in IFE-3 mRNPs, but excluded from IFE-1 mRNPs. These mRNAs also require IFE-1 for efficient translation. The findings support a model in which oocytes and embryos utilize the two eIF4Es for opposite purposes on critically regulated germline mRNAs. Careful colocalization of the eIF4Es with other germ granule components suggests an architecture in which GLH-1, PGL-1 and the IFEs are stratified to facilitate sequential interactions for mRNAs. Biochemical characterization demonstrates opposing yet cooperative roles for IFE-3 and IFE-1 to hand-off of translationally controlled mRNAs from the repressed to the activated state, respectively. The model involves eIF4E mRNPs shuttling mRNAs through nuclear pore-associated granules/condensates to cytoplasmic ribosomes.
Collapse
|
41
|
Mauro MS, Martin SL, Dumont J, Shirasu-Hiza M, Canman JC. Patterning, regulation, and role of FoxO/DAF-16 in the early embryo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.594029. [PMID: 38798632 PMCID: PMC11118310 DOI: 10.1101/2024.05.13.594029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Insulin resistance and diabetes are associated with many health issues including higher rates of birth defects and miscarriage during pregnancy. Because insulin resistance and diabetes are both associated with obesity, which also affects fertility, the role of insulin signaling itself in embryo development is not well understood. A key downstream target of the insulin/insulin-like growth factor signaling (IIS) pathway is the forkhead family transcription factor FoxO (DAF-16 in C. elegans ). Here, we used quantitative live imaging to measure the patterning of endogenously tagged FoxO/DAF-16 in the early worm embryo. In 2-4-cell stage embryos, FoxO/DAF-16 initially localized uniformly to all cell nuclei, then became dramatically enriched in germ precursor cell nuclei beginning at the 8-cell stage. This nuclear enrichment in early germ precursor cells required germ fate specification, PI3K (AGE-1)- and PTEN (DAF-18)-mediated phospholipid regulation, and the deubiquitylase USP7 (MATH-33), yet was unexpectedly insulin receptor (DAF-2)- and AKT-independent. Functional analysis revealed that FoxO/DAF-16 acts as a cell cycle pacer for early cleavage divisions-without FoxO/DAF-16 cell cycles were shorter than in controls, especially in germ lineage cells. These results reveal the germ lineage specific patterning, upstream regulation, and cell cycle role for FoxO/DAF-16 during early C. elegans embryogenesis.
Collapse
|
42
|
Spangler RK, Ashley GE, Braun K, Wruck D, Ramos-Coronado A, Ragle JM, Iesmantavicius V, Hess D, Partch CL, Großhans H, Ward JD. A conserved chronobiological complex times C. elegans development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593322. [PMID: 38766223 PMCID: PMC11100808 DOI: 10.1101/2024.05.09.593322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The mammalian PAS-domain protein PERIOD (PER) and its C. elegans orthologue LIN-42 have been proposed to constitute an evolutionary link between two distinct, circadian and developmental, timing systems. However, while the function of PER in animal circadian rhythms is well understood molecularly and mechanistically, this is not true for the function of LIN-42 in timing rhythmic development. Here, using targeted deletions, we find that the LIN-42 PAS domains are dispensable for the protein's function in timing molts. Instead, we observe arrhythmic molts upon deletion of a distinct sequence element, conserved with PER. We show that this element mediates stable binding to KIN-20, the C. elegans CK1δ/ε orthologue. We demonstrate that CK1δ phosphorylates LIN-42 and define two conserved helical motifs, CK1δ-binding domain A (CK1BD-A) and CK1BD-B, that have distinct roles in controlling CK1δ-binding and kinase activity in vitro. KIN-20 and the LIN-42 CK1BD are required for proper molting timing in vivo. These interactions mirror the central role of a stable circadian PER-CK1 complex in setting a robust ~24-hour period. Hence, our results establish LIN-42/PER - KIN-20/CK1δ/ε as a functionally conserved signaling module of two distinct chronobiological systems.
Collapse
Affiliation(s)
- Rebecca K Spangler
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Guinevere E Ashley
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Kathrin Braun
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Daniel Wruck
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Andrea Ramos-Coronado
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | | | - Daniel Hess
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Carrie L Partch
- Department of Chemistry and Biochemistry, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Helge Großhans
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
- University of Basel, 4002 Basel, Switzerland
| | - Jordan D Ward
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
43
|
Park S, Noblett N, Pitts L, Colavita A, Wehman AM, Jin Y, Chisholm AD. Dopey-dependent regulation of extracellular vesicles maintains neuronal morphology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.591898. [PMID: 38766017 PMCID: PMC11100700 DOI: 10.1101/2024.05.07.591898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Mature neurons maintain their distinctive morphology for extended periods in adult life. Compared to developmental neurite outgrowth, axon guidance, and target selection, relatively little is known of mechanisms that maintain mature neuron morphology. Loss of function in C. elegans DIP-2, a member of the conserved lipid metabolic regulator Dip2 family, results in progressive overgrowth of neurites in adults. We find that dip-2 mutants display specific genetic interactions with sax-2, the C. elegans ortholog of Drosophila Furry and mammalian FRY. Combined loss of DIP-2 and SAX-2 results in severe disruption of neuronal morphology maintenance accompanied by increased release of neuronal extracellular vesicles (EVs). By screening for suppressors of dip-2 sax-2 double mutant defects we identified gain-of-function (gf) mutations in the conserved Dopey family protein PAD-1 and its associated phospholipid flippase TAT-5/ATP9A. In dip-2 sax-2 double mutants carrying either pad-1(gf) or tat-5(gf) mutation, EV release is reduced and neuronal morphology across multiple neuron types is restored to largely normal. PAD-1(gf) acts cell autonomously in neurons. The domain containing pad-1(gf) is essential for PAD-1 function, and PAD-1(gf) protein displays increased association with the plasma membrane and inhibits EV release. Our findings uncover a novel functional network of DIP-2, SAX-2, PAD-1, and TAT-5 that maintains morphology of neurons and other types of cells, shedding light on the mechanistic basis of neurological disorders involving human orthologs of these genes.
Collapse
Affiliation(s)
- Seungmee Park
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Nathaniel Noblett
- Neuroscience Program, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Lauren Pitts
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | - Antonio Colavita
- Neuroscience Program, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Ann M Wehman
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Andrew D Chisholm
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
44
|
Torzone SK, Breen PC, Cohen NR, Simmons KN, Dowen RH. The TWK-26 potassium channel governs nutrient absorption in the C. elegans intestine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592787. [PMID: 38766028 PMCID: PMC11100751 DOI: 10.1101/2024.05.06.592787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Ion channels are necessary for proper water and nutrient absorption in the intestine, which supports cellular metabolism and organismal growth. While a role for Na + co-transporters and pumps in intestinal nutrient absorption is well defined, how individual K + uniporters function to maintain ion homeostasis is poorly understood. Using Caenorhabditis elegans , we show that a gain-of-function mutation in twk-26 , which encodes a two-pore domain K + ion channel orthologous to human KCNK3, facilitates nutrient absorption and suppresses the metabolic and developmental defects displayed by impaired intestinal MAP Kinase (MAPK) signaling. Mutations in drl-1 and flr-4, which encode two components of this MAPK pathway, cause severe growth defects, reduced lipid storage, and a dramatic increase in autophagic lysosomes, which mirror dietary restriction phenotypes. Additionally, these MAPK mutants display structural defects of the intestine and an impaired defecation motor program. We find that activation of TWK-26 reverses the dietary restriction-like state of the MAPK mutants by restoring intestinal nutrient absorption without correcting the intestinal bloating or defecation defects. This study provides unique insight into the mechanisms by which intestinal K + ion channels support intestinal metabolic homeostasis.
Collapse
|
45
|
VanDerMolen KR, Newman MA, Breen PC, Huff LA, Dowen RH. Non-cell-autonomous regulation of mTORC2 by Hedgehog signaling maintains lipid homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592795. [PMID: 38766075 PMCID: PMC11100691 DOI: 10.1101/2024.05.06.592795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Organisms must appropriately allocate energetic resources between essential cellular processes to maintain homeostasis and in turn, maximize fitness. The nutritional and homeostatic regulators of energy homeostasis have been studied in detail; however, how developmental signals might impinge on these pathways to govern cellular metabolism is poorly understood. Here, we identify a non-canonical role for Hedgehog (Hh), a classic regulator of development, in maintaining intestinal lipid homeostasis in C. elegans . We find that expression of two Hh ligands, GRD-3 and GRD-4, is controlled by the LIN-29/EGR transcription factor in the hypodermis, where the Hh secretion factor CHE-14/Dispatched also facilitates non-cell autonomous Hh signaling. We demonstrate, using C. elegans and mouse hepatocytes, that Hh metabolic regulation does not occur through the canonical Hh transcription factor, TRA-1/GLI, but rather through non-canonical signaling that engages mTOR Complex 2 (mTORC2) in the intestine. Hh mutants display impaired lipid homeostasis, including reduced lipoprotein synthesis and fat accumulation, decreased growth, and upregulation of autophagy factors, mimicking loss of mTORC2. Additionally, we found that Hh inhibits p38 MAPK signaling in parallel to mTORC2 activation and that both pathways act together to modulate of lipid homeostasis. Our findings show a non-canonical role for Hedgehog signaling in lipid metabolism via regulation of core homeostatic pathways and reveal a new mechanism by which developmental timing events govern metabolic decisions.
Collapse
|
46
|
Hayden AN, Brandel KL, Merlau PR, Vijayakumar P, Leptich EJ, Pietryk EW, Gaytan ES, Ni CW, Chao HT, Rosenfeld JA, Arey RN. Behavioral screening of conserved RNA-binding proteins reveals CEY-1/YBX RNA-binding protein dysfunction leads to impairments in memory and cognition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574402. [PMID: 38260399 PMCID: PMC10802296 DOI: 10.1101/2024.01.05.574402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
RNA-binding proteins (RBPs) regulate translation and plasticity which are required for memory. RBP dysfunction has been linked to a range of neurological disorders where cognitive impairments are a key symptom. However, of the 2,000 RBPs in the human genome, many are uncharacterized with regards to neurological phenotypes. To address this, we used the model organism C. elegans to assess the role of 20 conserved RBPs in memory. We identified eight previously uncharacterized memory regulators, three of which are in the C. elegans Y-Box (CEY) RBP family. Of these, we determined that cey-1 is the closest ortholog to the mammalian Y-Box (YBX) RBPs. We found that CEY-1 is both necessary in the nervous system for memory ability and sufficient to increase memory. Leveraging human datasets, we found both copy number variation losses and single nucleotide variants in YBX1 and YBX3 in individuals with neurological symptoms. We identified one predicted deleterious YBX3 variant of unknown significance, p.Asn127Tyr, in two individuals with neurological symptoms. Introducing this variant into endogenous cey-1 locus caused memory deficits in the worm. We further generated two humanized worm lines expressing human YBX3 or YBX1 at the cey-1 locus to test evolutionary conservation of YBXs in memory and the potential functional significance of the p.Asn127Tyr variant. Both YBX1/3 can functionally replace cey-1, and introduction of p.Asn127Tyr into the humanized YBX3 locus caused memory deficits. Our study highlights the worm as a model to reveal memory regulators and identifies YBX dysfunction as a potential new source of rare neurological disease.
Collapse
Affiliation(s)
- Ashley N Hayden
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Katie L Brandel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Paul R Merlau
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | | | - Emily J Leptich
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Edward W Pietryk
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
| | - Elizabeth S Gaytan
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Postbaccalaureate Research Education Program, Baylor College of Medicine, Houston, TX, 77030
| | - Connie W Ni
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Neuroscience, Rice University, Houston, TX 77005
| | - Hsiao-Tuan Chao
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, 77030
- Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, 77030
- McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, TX, 77030
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
- Baylor Genetics Laboratories, Houston, TX 77021
| | - Rachel N Arey
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| |
Collapse
|
47
|
Lamb H, Liro M, Myles K, Fernholz M, Anderson H, Rose LS. The Rac1 homolog CED-10 is a component of the MES-1/SRC-1 pathway for asymmetric division of the C. elegans EMS blastomere. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588162. [PMID: 38645195 PMCID: PMC11030239 DOI: 10.1101/2024.04.04.588162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Asymmetric cell division is essential for the creation of cell types with different identities and functions. The EMS blastomere of the four-cell Caenorhabditis elegans embryo undergoes an asymmetric division in response to partially redundant signaling pathways. One pathway involves a Wnt signal emanating from the neighboring P2 cell, while the other pathway is defined by the receptor-like MES-1 protein localized at the EMS/P2 cell contact, and the cytoplasmic kinase SRC-1. In response to these pathways, the EMS nuclear-centrosome complex rotates so that the spindle forms on the anterior-posterior axis; after division, the daughter cell contacting P2 becomes the endodermal precursor cell. Here we identify the Rac1 homolog, CED-10, as a new component of the MES-1/SRC-1 pathway. Loss of CED-10 affects both spindle positioning and endoderm specification. Although MES-1 is still present at the EMS/P2 contact in ced-10 embryos, SRC-1 dependent phosphorylation is reduced. These and other results suggest that CED-10 acts downstream of MES-1 and upstream of, or at the level of, SRC-1 activity. In addition, we find that the branched actin regulator ARX-2 is enriched at the EMS/P2 cell contact site, in a CED-10 dependent manner. Loss of ARX-2 results in spindle positioning defects, suggesting that CED-10 acts through branched actin to promote the asymmetric division of the EMS cell.
Collapse
Affiliation(s)
- Helen Lamb
- Department of Molecular and Cellular Biology, University of California, Davis One Shields Ave., Davis, CA 95616
| | - Małgorzata Liro
- Department of Molecular and Cellular Biology, University of California, Davis One Shields Ave., Davis, CA 95616
| | - Krista Myles
- Department of Molecular and Cellular Biology, University of California, Davis One Shields Ave., Davis, CA 95616
| | - McKenzi Fernholz
- Department of Molecular and Cellular Biology, University of California, Davis One Shields Ave., Davis, CA 95616
| | - Holly Anderson
- Department of Molecular and Cellular Biology, University of California, Davis One Shields Ave., Davis, CA 95616
| | - Lesilee S. Rose
- Department of Molecular and Cellular Biology, University of California, Davis One Shields Ave., Davis, CA 95616
| |
Collapse
|
48
|
Brar HK, Dey S, Singh P, Pande D, Ghosh-Roy A. Functional Recovery Associated with Dendrite Regeneration in PVD Neuron of Caenorhabditis elegans. eNeuro 2024; 11:ENEURO.0292-23.2024. [PMID: 38548333 PMCID: PMC7615967 DOI: 10.1523/eneuro.0292-23.2024] [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: 08/12/2023] [Revised: 02/18/2024] [Accepted: 03/03/2024] [Indexed: 05/02/2024] Open
Abstract
PVD neuron of Caenorhabditis elegans is a highly polarized cell with well-defined axonal, and dendritic compartments. PVD neuron operates in multiple sensory modalities including the control of both nociceptive touch sensation and body posture. Although both the axon and dendrites of this neuron show a regeneration response following laser-assisted injury, it is rather unclear how the behavior associated with this neuron is affected by the loss of these structures. It is also unclear whether neurite regrowth would lead to functional restoration in these neurons. Upon axotomy, using a femtosecond laser, we saw that harsh touch response was specifically affected leaving the body posture unperturbed. Subsequently, recovery in the touch response is highly correlated to the axon regrowth, which was dependent on DLK-1/MLK-1 MAP Kinase. Dendrotomy of both major and minor primary dendrites affected the wavelength and amplitude of sinusoidal movement without any apparent effect on harsh touch response. We further correlated the recovery in posture behavior to the type of dendrite regeneration events. We found that dendrite regeneration through the fusion and reconnection between the proximal and distal branches of the injured dendrite corresponded to improved recovery in posture. Our data revealed that the axons and dendrites of PVD neurons regulate the nociception and proprioception in worms, respectively. It also revealed that dendrite and axon regeneration lead to the restoration of these differential sensory modalities.
Collapse
Affiliation(s)
- Harjot Kaur Brar
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar 122052, Haryana, India
| | - Swagata Dey
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar 122052, Haryana, India
| | - Pallavi Singh
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar 122052, Haryana, India
| | - Devashish Pande
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar 122052, Haryana, India
| | - Anindya Ghosh-Roy
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar 122052, Haryana, India
| |
Collapse
|
49
|
Byrd DT, Han ZC, Piggott CA, Jin Y. PACS-1 variant protein is aberrantly localized in C. elegans model of PACS1/PACS2 syndromes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590644. [PMID: 38712144 PMCID: PMC11071410 DOI: 10.1101/2024.04.22.590644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
PACS (Phosphofurin Acidic Cluster Sorting Protein) proteins are known for their roles in sorting cargo proteins to organelles and can physically interact with WD40 repeat-containing protein WDR37. PACS1, PACS2, and WDR37 variants are associated with multisystemic syndromes and neurodevelopmental disorders characterized by intellectual disability, seizures, developmental delays, craniofacial abnormalities, and autism spectrum disorder. However, the effects of syndromic variants on function in vivo remains unknown. Here, we report the expression pattern of C. elegans orthologs of PACS and WDR37 and their interaction. We show that cePACS-1 and ceWDR-37 co-localize to somatic cytoplasm of many types of cells, and are mutually required for expression, supporting a conclusion that the intermolecular dependence of PACS1/PACS2/PACS-1 and WDR37/WDR-37 is evolutionarily conserved. We further show that editing in PACS1 and PACS2 variants in cePACS-1 changes protein localization in multiple cell types, including neurons. Moreover, expression of human PACS1 can functionally complement C. elegans PACS-1 in neurons, demonstrating conserved functions of the PACS-WDR37 axis in an invertebrate model system. Our findings reveal effects of human variants and suggest potential strategies to identify regulatory network components that may contribute to understanding molecular underpinnings of PACS/WDR37 syndromes.
Collapse
Affiliation(s)
- Dana T. Byrd
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093
| | - Ziyuan Christina Han
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093
| | - Christopher A. Piggott
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093
| |
Collapse
|
50
|
Wu Z, Cardona EA, Pierce JT. Non-apoptotic role of EGL-1 in exopher production and neuronal health in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590348. [PMID: 38712027 PMCID: PMC11071422 DOI: 10.1101/2024.04.19.590348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
While traditionally studied for their pro-apoptotic functions, recent research suggests BH3-only proteins also have non-apoptotic roles. Here, we find that EGL-1, the BH3-only protein in Caenorhabditis elegans, promotes the cell-autonomous production of exophers in adult neurons. Exophers are large, micron-scale vesicles that are ejected from the cell and contain cellular components such as mitochondria. EGL-1 facilitates exopher production potentially through regulation of mitochondrial dynamics. Moreover, an endogenous, low level of EGL-1 expression appears to benefit dendritic health. Our findings provide insights into the mechanistic role of BH3-only protein in mitochondrial dynamics, downstream exopher production, and ultimately neuronal health.
Collapse
Affiliation(s)
- Zheng Wu
- Department of Neuroscience, Center for Learning and Memory, Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX
| | - Eric A. Cardona
- Department of Neuroscience, Center for Learning and Memory, Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX
| | - Jonathan T. Pierce
- Department of Neuroscience, Center for Learning and Memory, Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX
| |
Collapse
|