Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Development ; 144(12): 2175-2186, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28634272

ABSTRACT

The major sperm protein domain (MSPd) has an extracellular signaling function implicated in amyotrophic lateral sclerosis. Secreted MSPds derived from the C. elegans VAPB homolog VPR-1 promote mitochondrial localization to actin-rich I-bands in body wall muscle. Here we show that the nervous system and germ line are key MSPd secretion tissues. MSPd signals are transduced through the CLR-1 Lar-like tyrosine phosphatase receptor. We show that CLR-1 is expressed throughout the muscle plasma membrane, where it is accessible to MSPd within the pseudocoelomic fluid. MSPd signaling is sufficient to remodel the muscle mitochondrial reticulum during adulthood. An RNAi suppressor screen identified survival of motor neuron 1 (SMN-1) as a downstream effector. SMN-1 acts in muscle, where it colocalizes at myofilaments with ARX-2, a component of the Arp2/3 actin-nucleation complex. Genetic studies suggest that SMN-1 promotes Arp2/3 activity important for localizing mitochondria to I-bands. Our results support the model that VAPB homologs are circulating hormones that pattern the striated muscle mitochondrial reticulum. This function is crucial in adults and requires SMN-1 in muscle, likely independent of its role in pre-mRNA splicing.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Membrane Proteins/metabolism , Muscle, Striated/growth & development , Muscle, Striated/metabolism , SMN Complex Proteins/metabolism , Actin-Related Protein 2/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Genes, Helminth , Germ Cells/metabolism , Humans , Larva/growth & development , Larva/metabolism , Male , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mitochondria, Muscle/metabolism , Motor Neurons/metabolism , Mutation , Protein Domains , RNA Interference , Receptor-Like Protein Tyrosine Phosphatases/genetics , Receptor-Like Protein Tyrosine Phosphatases/metabolism , SMN Complex Proteins/antagonists & inhibitors , SMN Complex Proteins/genetics , Sarcolemma/metabolism , Signal Transduction
2.
Development ; 144(12): 2187-2199, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28634273

ABSTRACT

VAMP/synaptobrevin-associated proteins (VAPs) contain an N-terminal major sperm protein domain (MSPd) that is associated with amyotrophic lateral sclerosis. VAPs have an intracellular housekeeping function, as well as an extracellular signaling function mediated by the secreted MSPd. Here we show that the C. elegans VAP homolog VPR-1 is essential for gonad development. vpr-1 null mutants are maternal effect sterile due to arrested gonadogenesis following embryo hatching. Somatic gonadal precursor cells and germ cells fail to proliferate fully and complete their respective differentiation programs. Maternal or zygotic vpr-1 expression is sufficient to induce gonadogenesis and fertility. Genetic mosaic and cell type-specific expression studies indicate that vpr-1 activity is important in the nervous system, germ line and intestine. VPR-1 acts in parallel to Notch signaling, a key regulator of germline stem cell proliferation and differentiation. Neuronal vpr-1 expression is sufficient for gonadogenesis induction during a limited time period shortly after hatching. These results support the model that the secreted VPR-1 MSPd acts at least in part on gonadal sheath cell precursors in L1 to early L2 stage hermaphrodites to permit gonadogenesis.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Gonads/growth & development , Gonads/metabolism , Membrane Proteins/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Differentiation , Female , Gene Knockout Techniques , Genome, Helminth , Germ Cells/cytology , Germ Cells/metabolism , Intestinal Mucosa/metabolism , Intestines/growth & development , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Models, Biological , Mosaicism , Neurogenesis , Organogenesis , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction
3.
Science ; 344(6185): 754-7, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24833393

ABSTRACT

Environmental exposures affect gamete function and fertility, but the mechanisms are poorly understood. Here, we show that pheromones sensed by ciliated neurons in the Caenorhabditis elegans nose alter the lipid microenvironment within the oviduct, thereby affecting sperm motility. In favorable environments, pheromone-responsive sensory neurons secrete a transforming growth factor-ß ligand called DAF-7, which acts as a neuroendocrine factor that stimulates prostaglandin-endoperoxide synthase [cyclooxygenase (Cox)]-independent prostaglandin synthesis in the ovary. Oocytes secrete F-class prostaglandins that guide sperm toward them. These prostaglandins are also synthesized in Cox knockout mice, raising the possibility that similar mechanisms exist in other animals. Our data indicate that environmental cues perceived by the female nervous system affect sperm function.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Fertilization , Neurons, Afferent/physiology , Pheromones/physiology , Sperm Motility , Spermatozoa/physiology , Transforming Growth Factor beta/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Environmental Exposure , Female , Male , Neurosecretory Systems/physiology , Oocytes/metabolism , Oocytes/physiology , Ovum/metabolism , Ovum/physiology , Perception , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/biosynthesis , Transforming Growth Factor beta/genetics
4.
PLoS Genet ; 9(9): e1003738, 2013.
Article in English | MEDLINE | ID: mdl-24039594

ABSTRACT

Mutations in VAPB/ALS8 are associated with amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), two motor neuron diseases that often include alterations in energy metabolism. We have shown that C. elegans and Drosophila neurons secrete a cleavage product of VAPB, the N-terminal major sperm protein domain (vMSP). Secreted vMSPs signal through Roundabout and Lar-like receptors expressed on striated muscle. The muscle signaling pathway localizes mitochondria to myofilaments, alters their fission/fusion balance, and promotes energy production. Here, we show that neuronal loss of the C. elegans VAPB homolog triggers metabolic alterations that appear to compensate for muscle mitochondrial dysfunction. When vMSP levels drop, cytoskeletal or mitochondrial abnormalities in muscle induce elevated DAF-16, the Forkhead Box O (FoxO) homolog, transcription factor activity. DAF-16 promotes muscle triacylglycerol accumulation, increases ATP levels in adults, and extends lifespan, despite reduced muscle mitochondria electron transport chain activity. Finally, Vapb knock-out mice exhibit abnormal muscular triacylglycerol levels and FoxO target gene transcriptional responses to fasting and refeeding. Our data indicate that impaired vMSP signaling to striated muscle alters FoxO activity, which affects energy metabolism. Abnormalities in energy metabolism of ALS patients may thus constitute a compensatory mechanism counterbalancing skeletal muscle mitochondrial dysfunction.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Energy Metabolism , Membrane Proteins/genetics , Muscle, Striated/metabolism , Muscular Atrophy, Spinal/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Drosophila/metabolism , Forkhead Transcription Factors , Humans , Ligands , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondria/pathology , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Signal Transduction , Transcription Factors/metabolism , Vesicular Transport Proteins
5.
Dev Cell ; 22(2): 348-62, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22264801

ABSTRACT

VIDEO ABSTRACT: The VAPB/ALS8 major sperm protein domain (vMSP) is implicated in amyotrophic lateral sclerosis and spinal muscular atrophy, yet its function in the nervous system is not well understood. In Caenorhabditis elegans and Drosophila, the vMSP is cleaved from its transmembrane anchor and secreted in a cell type-specific fashion. We show that vMSPs secreted by neurons act on Lar-like protein-tyrosine phosphatase and Roundabout growth cone guidance receptors expressed in striated muscle. This signaling pathway promotes Arp2/3-dependent actin remodeling and mitochondrial localization to actin-rich muscle I-bands. C. elegans VAPB mutants have mitochondrial localization, morphology, mobility, and fission/fusion defects that are suppressed by Lar-like receptor or Arp2/3 inactivation. Hence, growth cone guidance receptor pathways that remodel the actin cytoskeleton have unanticipated effects on mitochondrial dynamics. We propose that neurons secrete vMSPs to promote striated muscle energy production and metabolism, in part through the regulation of mitochondrial localization and function.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Growth Cones/metabolism , Helminth Proteins/metabolism , Mitochondria/metabolism , Muscle, Striated/metabolism , Neurons/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Fluorescence , Helminth Proteins/genetics , Mitochondria/pathology , Muscle, Striated/cytology , Neurons/cytology , Protein Structure, Tertiary , Signal Transduction , Transgenes/physiology
6.
Dev Cell ; 19(6): 858-71, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21145501

ABSTRACT

Abnormalities in insulin/IGF-1 signaling are associated with infertility, but the molecular mechanisms are not well understood. Here we use liquid chromatography with electrospray ionization tandem mass spectrometry to show that the C. elegans insulin/FOXO pathway regulates the metabolism of locally acting lipid hormones called prostaglandins. C. elegans prostaglandins are synthesized without prostaglandin G/H synthase homologs, the targets of nonsteroidal anti-inflammatory drugs. Our results support the model that insulin signaling promotes the conversion of oocyte polyunsaturated fatty acids (PUFAs) into F-series prostaglandins that guide sperm to the fertilization site. Reduction in insulin signaling activates DAF-16/FOXO, which represses the transcription of germline and intestinal genes required to deliver PUFAs to oocytes in lipoprotein complexes. Nutritional and neuroendocrine cues target this mechanism to control prostaglandin metabolism and reproductive output. Prostaglandins may be conserved sperm guidance factors and widespread downstream effectors of insulin actions that influence both reproductive and nonreproductive processes.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Forkhead Transcription Factors/physiology , Insulin/physiology , Ovary/physiology , Prostaglandins F/physiology , Animals , Animals, Genetically Modified , Base Sequence , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , DNA Primers/genetics , Fatty Acids, Unsaturated/metabolism , Female , Genes, Helminth , Male , Models, Biological , Mutation , Oocytes/physiology , Receptor, Insulin/genetics , Receptor, Insulin/physiology , Reproduction/physiology , Signal Transduction , Spectrometry, Mass, Electrospray Ionization , Sperm Motility/physiology , Tandem Mass Spectrometry , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL