Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.005
Filter
1.
Methods Mol Biol ; 2792: 265-275, 2024.
Article in English | MEDLINE | ID: mdl-38861094

ABSTRACT

Eukaryotic cells are compartmentalized by membrane-bounded organelles to ensure that specific biochemical reactions and cellular functions occur in a spatially restricted manner. The subcellular localization of proteins is largely determined by their intrinsic targeting signals, which are mainly constituted by short peptides. A complete organelle targeting signal may contain a core signal (CoreS) as well as auxiliary signals (AuxiS). However, the AuxiS is often not as well characterized as the CoreS. Peroxisomes house many key steps in photorespiration, besides other crucial functions in plants. Peroxisome targeting signal type 1 (PTS1), which is carried by most peroxisome matrix proteins, was initially recognized as a C-terminal tripeptide with a "canonical" consensus of [S/A]-[K/R]-[L/M]. Many studies have shown the existence of auxiliary targeting signals upstream of PTS1, but systematic characterizations are lacking. Here, we designed an analytical strategy to characterize the auxiliary targeting signals for plant peroxisomes using large datasets and statistics followed by experimental validations. This method may also be applied to deciphering the auxiliary targeting signals for other organelles, whose organellar targeting depends on a core peptide with assistance from a nearby auxiliary signal.


Subject(s)
Computational Biology , Peroxisomes , Peroxisomes/metabolism , Computational Biology/methods , Protein Transport , Peroxisomal Targeting Signals , Protein Sorting Signals , Plant Proteins/metabolism , Plant Proteins/genetics , Databases, Protein , Amino Acid Sequence
2.
Cell Mol Biol Lett ; 29(1): 85, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834954

ABSTRACT

The molecular basis for bulk autophagy activation due to a deficiency in essential nutrients such as carbohydrates, amino acids, and nitrogen is well understood. Given autophagy functions to reduce surplus to compensate for scarcity, it theoretically possesses the capability to selectively degrade specific substrates to meet distinct metabolic demands. However, direct evidence is still lacking that substantiates the idea that autophagy selectively targets specific substrates (known as selective autophagy) to address particular nutritional needs. Recently, Gross et al. found that during phosphate starvation (P-S), rather than nitrogen starvation (N-S), yeasts selectively eliminate peroxisomes by dynamically altering the composition of the Atg1/ULK kinase complex (AKC) to adapt to P-S. This study elucidates how the metabolite sensor Pho81 flexibly interacts with AKC and guides selective autophagic clearance of peroxisomes during P-S, providing novel insights into the metabolic contribution of autophagy to special nutritional needs.


Subject(s)
Autophagy , Phosphates , Saccharomyces cerevisiae Proteins , Phosphates/metabolism , Phosphates/deficiency , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Peroxisomes/metabolism , Saccharomyces cerevisiae/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Kinases
3.
Nat Commun ; 15(1): 4314, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773129

ABSTRACT

Peroxisomes are eukaryotic organelles that are essential for multiple metabolic pathways, including fatty acid oxidation, degradation of amino acids, and biosynthesis of ether lipids. Consequently, peroxisome dysfunction leads to pediatric-onset neurodegenerative conditions, including Peroxisome Biogenesis Disorders (PBD). Due to the dynamic, tissue-specific, and context-dependent nature of their biogenesis and function, live cell imaging of peroxisomes is essential for studying peroxisome regulation, as well as for the diagnosis of PBD-linked abnormalities. However, the peroxisomal imaging toolkit is lacking in many respects, with no reporters for substrate import, nor cell-permeable probes that could stain dysfunctional peroxisomes. Here we report that the BODIPY-C12 fluorescent fatty acid probe stains functional and dysfunctional peroxisomes in live mammalian cells. We then go on to improve BODIPY-C12, generating peroxisome-specific reagents, PeroxiSPY650 and PeroxiSPY555. These probes combine high peroxisome specificity, bright fluorescence in the red and far-red spectrum, and fast non-cytotoxic staining, making them ideal tools for live cell, whole organism, or tissue imaging of peroxisomes. Finally, we demonstrate that PeroxiSPY enables diagnosis of peroxisome abnormalities in the PBD CRISPR/Cas9 cell models and patient-derived cell lines.


Subject(s)
Boron Compounds , Fatty Acids , Fluorescent Dyes , Peroxisomal Disorders , Peroxisomes , Peroxisomes/metabolism , Humans , Fatty Acids/metabolism , Fluorescent Dyes/chemistry , Boron Compounds/chemistry , Peroxisomal Disorders/metabolism , Animals
4.
Int J Biol Macromol ; 270(Pt 2): 132227, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38734339

ABSTRACT

Fusarium crown rot, caused by Fusarium pseudograminearum, is a devastating disease affecting the yield and quality of cereal crops. Peroxisomes are single-membrane organelles that play a critical role in various biological processes in eukaryotic cells. To functionally characterise peroxisome biosynthetic receptor proteins FpPEX5 and FpPEX7 in F. pseudograminearum, we constructed deletion mutants, ΔFpPEX5 and ΔFpPEX7, and complementary strains, ΔFpPEX5-C and ΔFpPEX7-C, and analysed the functions of FpPEX5 and FpPEX7 proteins using various phenotypic observations. The deletion of FpPEX5 and FpPEX7 resulted in a significant deficiency in mycelial growth and conidiation and blocked the peroxisomal targeting signal 1 and peroxisomal targeting signal 2 pathways, which are involved in peroxisomal matrix protein transport, increasing the accumulation of lipid droplets and reactive oxygen species. The deletion of FpPEX5 and FpPEX7 may reduce the formation of toxigenic bodies and decrease the pathogenicity of F. pseudograminearum. These results indicate that FpPEX5 and FpPEX7 play vital roles in the growth, asexual reproduction, virulence, and fatty acid utilisation of F. pseudograminearum. This study provides a theoretical basis for controlling stem rot in wheat.


Subject(s)
Fungal Proteins , Fusarium , Peroxisomes , Fusarium/pathogenicity , Fusarium/genetics , Fusarium/metabolism , Fusarium/growth & development , Fungal Proteins/genetics , Fungal Proteins/metabolism , Virulence/genetics , Peroxisomes/metabolism , Peroxisomes/genetics , Trichothecenes/metabolism , Plant Diseases/microbiology , Spores, Fungal/growth & development , Triticum/microbiology , Reactive Oxygen Species/metabolism , Peroxisome-Targeting Signal 1 Receptor/genetics , Peroxisome-Targeting Signal 1 Receptor/metabolism , Gene Deletion , Gene Expression Regulation, Fungal , Peroxisomal Targeting Signal 2 Receptor , Mycelium/growth & development , Mycelium/metabolism
5.
Biochim Biophys Acta Mol Cell Res ; 1871(6): 119754, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38762172

ABSTRACT

Peroxisome biogenesis disorders are caused by pathogenic variants in genes involved in biogenesis and maintenance of peroxisomes. However, mitochondria are also often affected in these diseases. Peroxisomal membrane proteins, including PEX14, have been found to mislocalise to mitochondria in cells lacking peroxisomes. Recent studies indicated that this mislocalisation contributes to mitochondrial abnormalities in PEX3-deficient patient fibroblasts cells. Here, we studied whether mitochondrial morphology is also affected in PEX3-deficient HEK293 cells and whether PEX14 mislocalises to mitochondria in these cells. Using high-resolution imaging techniques, we show that although endogenous PEX14 mislocalises to mitochondria, mitochondrial morphology was normal in PEX3-KO HEK293 cells. However, we discovered that overexpression of tagged PEX14 in wild-type HEK293 cells resulted in its mitochondrial localisation, accompanied by altered mitochondrial morphology. Our data indicate that overexpression of tagged PEX14 alone directly or indirectly cause mitochondrial abnormalities in cells containing peroxisomes.


Subject(s)
Membrane Proteins , Mitochondria , Peroxisomes , Humans , Mitochondria/metabolism , Mitochondria/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , HEK293 Cells , Peroxisomes/metabolism , Peroxisomes/genetics , Peroxins/metabolism , Peroxins/genetics , Protein Transport , Lipoproteins , Repressor Proteins
7.
Int J Biochem Cell Biol ; 172: 106585, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734232

ABSTRACT

Tamoxifen is an estrogen receptor modulator that has been reported to alleviate hepatic lipid accumulation in mice, but the mechanism is still unclear. Peroxisome fatty acid ß-oxidation is the main metabolic pathway for the overload of long-chain fatty acids. As long-chain fatty acids are a cause of hepatic lipid accumulation, the activation of peroxisome fatty acid ß-oxidation might be a novel therapeutic strategy for metabolic associated fatty liver disease. In this study, we investigated the mechanism of tamoxifen against hepatic lipid accumulation based on the activation of peroxisome fatty acid ß-oxidation. Tamoxifen reduced liver long-chain fatty acids and relieved hepatic lipid accumulation in high fat diet mice without sex difference. In vitro, tamoxifen protected primary hepatocytes against palmitic acid-induced lipotoxicity. Mechanistically, the RNA-sequence of hepatocytes isolated from the liver revealed that peroxisome fatty acid ß-oxidation was activated by tamoxifen. Protein and mRNA expression of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase were significantly increased in vivo and in vitro. Small interfering RNA enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase in primary hepatocytes abolished the therapeutic effects of tamoxifen in lipid accumulation. In conclusion, our results indicated that tamoxifen could relieve hepatic lipid accumulation in high fat diet mice based on the activation of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase-mediated peroxisome fatty acids ß-oxidation.


Subject(s)
Enoyl-CoA Hydratase , Hepatocytes , Lipid Metabolism , Liver , Mice, Inbred C57BL , Oxidation-Reduction , Peroxisomes , Tamoxifen , Animals , Tamoxifen/pharmacology , Mice , Lipid Metabolism/drug effects , Liver/metabolism , Liver/drug effects , Hepatocytes/metabolism , Hepatocytes/drug effects , Oxidation-Reduction/drug effects , Male , Peroxisomes/metabolism , Peroxisomes/drug effects , Enoyl-CoA Hydratase/metabolism , Enoyl-CoA Hydratase/genetics , Up-Regulation/drug effects , Diet, High-Fat/adverse effects , Female , Fatty Acids/metabolism
8.
PLoS One ; 19(5): e0298274, 2024.
Article in English | MEDLINE | ID: mdl-38753762

ABSTRACT

The membrane peroxisomal proteins PEX11, play a crucial role in peroxisome proliferation by regulating elongation, membrane constriction, and fission of pre-existing peroxisomes. In this study, we evaluated the function of PEX11B gene in neural differentiation of human embryonic stem cell (hESC) by inducing shRNAi-mediated knockdown of PEX11B expression. Our results demonstrate that loss of PEX11B expression led to a significant decrease in the expression of peroxisomal-related genes including ACOX1, PMP70, PEX1, and PEX7, as well as neural tube-like structures and neuronal markers. Inhibition of SIRT1 using pharmacological agents counteracted the effects of PEX11B knockdown, resulting in a relative increase in PEX11B expression and an increase in differentiated neural tube-like structures. However, the neuroprotective effects of SIRT1 were eliminated by PPAR inhibition, indicating that PPARÉ£ may mediate the interaction between PEX11B and SIRT1. Our findings suggest that both SIRT1 and PPARÉ£ have neuroprotective effects, and also this study provides the first indication for a potential interaction between PEX11B, SIRT1, and PPARÉ£ during hESC neural differentiation.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells , Membrane Proteins , PPAR gamma , Sirtuin 1 , Humans , Sirtuin 1/metabolism , Sirtuin 1/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , Cell Differentiation/drug effects , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Neurons/metabolism , Neurons/cytology , Neurons/drug effects , Cell Line , Peroxisomes/metabolism
9.
Free Radic Biol Med ; 221: 81-88, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38762061

ABSTRACT

Androgen receptor (AR)-targeting therapy induces oxidative stress in prostate cancer. However, the mechanism of oxidative stress induction by AR-targeting therapy remains unclear. This study investigated the mechanism of oxidative stress induction by AR-targeting therapy, with the aim to develop novel therapeutics targeting oxidative stress induced by AR-targeting therapy. Intracellular reactive oxygen species (ROS) was examined by fluorescence microscopy and flow cytometry analysis. The effects of silencing gene expression and small molecule inhibitors on gene expression and cytotoxic effects were examined by quantitative real-time PCR and cell proliferation assay. ROS induced by androgen depletion co-localized with peroxisomes in prostate cancer cells. Among peroxisome-related genes, PPARA was commonly induced by AR inhibition and involved in ROS production via PKC signaling. Inhibition of PPARα by specific siRNA and a small molecule inhibitor suppressed cell proliferation and increased cellular sensitivity to the antiandrogen enzalutamide in prostate cancer cells. This study revealed a novel pathway by which AR inhibition induced intracellular ROS mainly in peroxisomes through PPARα activation in prostate cancer. This pathway is a promising target for the development of novel therapeutics for prostate cancer in combination with AR-targeting therapy such as antiandrogen enzalutamide.


Subject(s)
Benzamides , Cell Proliferation , Drug Resistance, Neoplasm , Nitriles , Oxidative Stress , PPAR alpha , Peroxisomes , Phenylthiohydantoin , Prostatic Neoplasms , Reactive Oxygen Species , Receptors, Androgen , Male , Humans , Phenylthiohydantoin/pharmacology , Nitriles/pharmacology , Peroxisomes/metabolism , Peroxisomes/drug effects , Oxidative Stress/drug effects , Drug Resistance, Neoplasm/drug effects , Benzamides/pharmacology , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Reactive Oxygen Species/metabolism , PPAR alpha/metabolism , PPAR alpha/genetics , Cell Proliferation/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Signal Transduction/drug effects , Androgen Receptor Antagonists/pharmacology , RNA, Small Interfering/genetics
10.
J Cell Sci ; 137(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38752931

ABSTRACT

Peroxisomes are highly plastic organelles that are involved in several metabolic processes, including fatty acid oxidation, ether lipid synthesis and redox homeostasis. Their abundance and activity are dynamically regulated in response to nutrient availability and cellular stress. Damaged or superfluous peroxisomes are removed mainly by pexophagy, the selective autophagy of peroxisomes induced by ubiquitylation of peroxisomal membrane proteins or ubiquitin-independent processes. Dysregulated pexophagy impairs peroxisome homeostasis and has been linked to the development of various human diseases. Despite many recent insights into mammalian pexophagy, our understanding of this process is still limited compared to our understanding of pexophagy in yeast. In this Cell Science at a Glance article and the accompanying poster, we summarize current knowledge on the control of mammalian pexophagy and highlight which aspects require further attention. We also discuss the role of ubiquitylation in pexophagy and describe the ubiquitin machinery involved in regulating signals for the recruitment of phagophores to peroxisomes.


Subject(s)
Peroxisomes , Ubiquitination , Peroxisomes/metabolism , Humans , Animals , Autophagy , Macroautophagy , Mammals/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics
11.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119742, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38702017

ABSTRACT

Peroxisomes are ubiquitous cell organelles involved in various metabolic pathways. In order to properly function, several cofactors, substrates and products of peroxisomal enzymes need to pass the organellar membrane. So far only a few transporter proteins have been identified. We analysed peroxisomal membrane fractions purified from the yeast Hansenula polymorpha by untargeted label-free quantitation mass spectrometry. As expected, several known peroxisome-associated proteins were enriched in the peroxisomal membrane fraction. In addition, several other proteins were enriched, including mitochondrial transport proteins. Localization studies revealed that one of them, the mitochondrial phosphate carrier Mir1, has a dual localization on mitochondria and peroxisomes. To better understand the molecular mechanisms of dual sorting, we localized Mir1 in cells lacking Pex3 or Pex19, two peroxins that play a role in targeting of peroxisomal membrane proteins. In these cells Mir1 only localized to mitochondria, indicating that Pex3 and Pex19 are required to sort Mir1 to peroxisomes. Analysis of the localization of truncated versions of Mir1 in wild-type H. polymorpha cells revealed that most of them localized to mitochondria, but only one, consisting of the transmembrane domains 3-6, was peroxisomal. Peroxisomal localization of this construct was lost in a MIR1 deletion strain, indicating that full-length Mir1 was required for the localization of the truncated protein to peroxisomes. Our data suggest that only full-length Mir1 sorts to peroxisomes, while Mir1 contains multiple regions with mitochondrial sorting information. Data are available via ProteomeXchange with identifier PXD050324.


Subject(s)
Fungal Proteins , Mitochondria , Peroxisomes , Pichia , Peroxisomes/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Fungal Proteins/metabolism , Fungal Proteins/genetics , Pichia/metabolism , Pichia/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Peroxins/metabolism , Peroxins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Protein Transport
12.
Insect Biochem Mol Biol ; 170: 104139, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815735

ABSTRACT

Peroxisomes are ubiquitous cellular organelles participating in a variety of critical metabolic reactions. PEX14 is an essential peroxin responsible for peroxisome biogenesis. In this study, we identified the human PEX14 homolog in the brown planthopper, Nilaparvata lugens (Hemiptera: Delphacidae). N. lugens PEX14 (NlPEX14) showed significant topological similarity to its human counterpart. It is expressed throughout all developmental stages, with the highest expression observed in adult insects. Down-regulation of NlPEX14 through injection of NlPEX14-specific double-strand RNA impaired nymphal development. Moreover, females subjected to dsNlPEX14 treatment exhibited a significantly reduced lifespan. Additionally, we found abnormal ovarian development and a significant decrease in the number of eggs laid in NlPEX14-downregulated females. Further experiments support that the shortening of lifespan and the decrease in female fecundity can be attributed, at least partially, to the accumulation of fatty acids and reduced expression of vitellogenin. Together, our study reveals an indispensable function of NlPEX14 for insect reproduction and establishes a causal connection between the phenotypes and peroxisome biogenesis, shedding light on the importance of peroxisomes in female fecundity.


Subject(s)
Fertility , Hemiptera , Insect Proteins , Animals , Hemiptera/genetics , Hemiptera/metabolism , Hemiptera/physiology , Hemiptera/growth & development , Female , Insect Proteins/metabolism , Insect Proteins/genetics , Peroxisomes/metabolism , Longevity , Nymph/growth & development , Nymph/metabolism , Nymph/genetics , Peroxins/metabolism , Peroxins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Vitellogenins/metabolism , Vitellogenins/genetics
13.
Cell Mol Life Sci ; 81(1): 190, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649521

ABSTRACT

The high-protein diet (HPD) has emerged as a potent dietary approach to curb obesity. Peroxisome, a highly malleable organelle, adapts to nutritional changes to maintain homeostasis by remodeling its structure, composition, and quantity. However, the impact of HPD on peroxisomes and the underlying mechanism remains elusive. Using Drosophila melanogaster as a model system, we discovered that HPD specifically increases peroxisome levels within the adipose tissues. This HPD-induced peroxisome elevation is attributed to cysteine and methionine by triggering the expression of CG33474, a fly homolog of mammalian PEX11G. Both the overexpression of Drosophila CG33474 and human PEX11G result in increased peroxisome size. In addition, cysteine and methionine diets both reduce lipid contents, a process that depends on the presence of CG33474. Furthermore, CG33474 stimulates the breakdown of neutral lipids in a cell-autonomous manner. Moreover, the expression of CG33474 triggered by cysteine and methionine requires TOR signaling. Finally, we found that CG33474 promotes inter-organelle contacts between peroxisomes and lipid droplets (LDs), which might be a potential mechanism for CG33474-induced fat loss. In summary, our findings demonstrate that CG33474/PEX11G may serve as an essential molecular bridge linking HPD to peroxisome dynamics and lipid metabolism.


Subject(s)
Adipose Tissue , Cysteine , Drosophila Proteins , Drosophila melanogaster , Methionine , Peroxisomes , Animals , Methionine/metabolism , Peroxisomes/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Cysteine/metabolism , Adipose Tissue/metabolism , Humans , Lipid Metabolism , Lipid Droplets/metabolism , Signal Transduction , Diet
14.
Nat Commun ; 15(1): 3317, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38632234

ABSTRACT

Import of proteins into peroxisomes depends on PEX5, PEX13 and PEX14. By combining biochemical methods and structural biology, we show that the C-terminal SH3 domain of PEX13 mediates intramolecular interactions with a proximal FxxxF motif. The SH3 domain also binds WxxxF peptide motifs in the import receptor PEX5, demonstrating evolutionary conservation of such interactions from yeast to human. Strikingly, intramolecular interaction of the PEX13 FxxxF motif regulates binding of PEX5 WxxxF/Y motifs to the PEX13 SH3 domain. Crystal structures reveal how FxxxF and WxxxF/Y motifs are recognized by a non-canonical surface on the SH3 domain. The PEX13 FxxxF motif also mediates binding to PEX14. Surprisingly, the potential PxxP binding surface of the SH3 domain does not recognize PEX14 PxxP motifs, distinct from its yeast ortholog. Our data show that the dynamic network of PEX13 interactions with PEX5 and PEX14, mediated by diaromatic peptide motifs, modulates peroxisomal matrix import.


Subject(s)
Membrane Proteins , Protein Transport , Saccharomyces cerevisiae Proteins , src Homology Domains , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Peptides/chemistry , Peroxisome-Targeting Signal 1 Receptor/metabolism , Peroxisomes/metabolism , Protein Binding , Protein Transport/genetics , Protein Transport/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , src Homology Domains/genetics , src Homology Domains/physiology
15.
Biotechnol Bioeng ; 121(7): 2091-2105, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38568751

ABSTRACT

Peroxisomal compartmentalization has emerged as a highly promising strategy for reconstituting intricate metabolic pathways. In recent years, significant progress has been made in the peroxisomes through harnessing precursor pools, circumventing metabolic crosstalk, and minimizing the cytotoxicity of exogenous pathways. However, it is important to note that in methylotrophic yeasts (e.g. Pichia pastoris), the abundance and protein composition of peroxisomes are highly variable, particularly when peroxisome proliferation is induced by specific carbon sources. The intricate subcellular localization of native proteins, the variability of peroxisomal metabolic pathways, and the lack of systematic characterization of peroxisome targeting signals have limited the applications of peroxisomal compartmentalization in P. pastoris. Accordingly, this study established a high-throughput screening method based on ß-carotene biosynthetic pathway to evaluate the targeting efficiency of PTS1s (Peroxisome Targeting Signal Type 1) in P. pastoris. First, 25 putative endogenous PTS1s were characterized and 3 PTS1s with high targeting efficiency were identified. Then, directed evolution of PTS1s was performed by constructing two PTS1 mutant libraries, and a total of 51 PTS1s (29 classical and 22 noncanonical PTS1s) with presumably higher peroxisomal targeting efficiency were identified, part of which were further characterized via confocal microscope. Finally, the newly identified PTS1s were employed for peroxisomal compartmentalization of the geraniol biosynthetic pathway, resulting in more than 30% increase in the titer of monoterpene compared with when the pathway was localized to the cytosol. The present study expands the synthetic biology toolkit and lays a solid foundation for peroxisomal compartmentalization in P. pastoris.


Subject(s)
Metabolic Engineering , Peroxisomes , Peroxisomes/metabolism , Peroxisomes/genetics , Metabolic Engineering/methods , Peroxisomal Targeting Signals/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Pichia/genetics , Pichia/metabolism , Saccharomycetales/genetics , Saccharomycetales/metabolism
16.
Dev Cell ; 59(11): 1363-1378.e4, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38579719

ABSTRACT

The mechanism underlying the ability of rice to germinate underwater is a largely enigmatic but key research question highly relevant to rice cultivation. Moreover, although rice is known to accumulate salicylic acid (SA), SA biosynthesis is poorly defined, and its role in underwater germination is unknown. It is also unclear whether peroxisomes, organelles essential to oilseed germination and rice SA accumulation, play a role in rice germination. Here, we show that submerged imbibition of rice seeds induces SA accumulation to promote germination in submergence. Two submergence-induced peroxisomal Oryza sativa cinnamate:CoA ligases (OsCNLs) are required for this SA accumulation. SA exerts this germination-promoting function by inducing indole-acetic acid (IAA) catabolism through the IAA-amino acid conjugating enzyme GH3. The metabolic cascade we identified may potentially be adopted in agriculture to improve the underwater germination of submergence-intolerant rice varieties. SA pretreatment is also a promising strategy to improve submerged rice germination in the field.


Subject(s)
Germination , Oryza , Peroxisomes , Plant Growth Regulators , Plant Proteins , Oryza/metabolism , Oryza/growth & development , Germination/physiology , Peroxisomes/metabolism , Plant Growth Regulators/metabolism , Plant Proteins/metabolism , Gene Expression Regulation, Plant , Coenzyme A Ligases/metabolism , Indoleacetic Acids/metabolism , Seeds/metabolism , Seeds/growth & development , Salicylic Acid/metabolism , Cinnamates/metabolism
17.
Biol Open ; 13(5)2024 May 15.
Article in English | MEDLINE | ID: mdl-38682287

ABSTRACT

Pex23 family proteins localize to the endoplasmic reticulum and play a role in peroxisome and lipid body formation. The yeast Hansenula polymorpha contains four members: Pex23, Pex24, Pex29 and Pex32. We previously showed that loss of Pex24 or Pex32 results in severe peroxisomal defects, caused by reduced peroxisome-endoplasmic reticulum contact sites. We now analyzed the effect of the absence of all four Pex23 family proteins on other cell organelles. Vacuoles were normal in all four deletion strains. The number of lipid droplets was reduced in pex23 and pex29, but not in pex24 and pex32 cells, indicating that peroxisome and lipid droplet formation require different Pex23 family proteins in H. polymorpha. In pex23 and pex29 cells mitochondria were fragmented and clustered accompanied by reduced levels of the fusion protein Fzo1. Deletion of DNM1 suppressed the morphological phenotype of pex23 and pex29 cells, suggesting that mitochondrial fusion is affected. pex23 and pex29 cells showed retarded growth and reduced mitochondrial activities. The growth defect was partially suppressed by DNM1 deletion as well as by an artificial mitochondrion-endoplasmic reticulum tether. Hence, the absence of Pex23 family proteins may influence mitochondrion-endoplasmic reticulum contact sites.


Subject(s)
Endoplasmic Reticulum , Mitochondria , Peroxins , Peroxisomes , Pichia , Mitochondria/metabolism , Endoplasmic Reticulum/metabolism , Pichia/metabolism , Pichia/genetics , Peroxins/metabolism , Peroxins/genetics , Peroxisomes/metabolism , Gene Deletion , Fungal Proteins/metabolism , Fungal Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Vacuoles/metabolism , Phenotype
18.
PLoS Biol ; 22(4): e3002602, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38669296

ABSTRACT

Mitofusins are large GTPases that trigger fusion of mitochondrial outer membranes. Similarly to the human mitofusin Mfn2, which also tethers mitochondria to the endoplasmic reticulum (ER), the yeast mitofusin Fzo1 stimulates contacts between Peroxisomes and Mitochondria when overexpressed. Yet, the physiological significance and function of these "PerMit" contacts remain unknown. Here, we demonstrate that Fzo1 naturally localizes to peroxisomes and promotes PerMit contacts in physiological conditions. These contacts are regulated through co-modulation of Fzo1 levels by the ubiquitin-proteasome system (UPS) and by the desaturation status of fatty acids (FAs). Contacts decrease under low FA desaturation but reach a maximum during high FA desaturation. High-throughput genetic screening combined with high-resolution cellular imaging reveal that Fzo1-mediated PerMit contacts favor the transit of peroxisomal citrate into mitochondria. In turn, citrate enters the TCA cycle to stimulate the mitochondrial membrane potential and maintain efficient mitochondrial fusion upon high FA desaturation. These findings thus unravel a mechanism by which inter-organelle contacts safeguard mitochondrial fusion.


Subject(s)
Mitochondria , Mitochondrial Dynamics , Peroxisomes , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Peroxisomes/metabolism , Mitochondrial Dynamics/physiology , Mitochondria/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Fatty Acids/metabolism , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Citric Acid Cycle , Membrane Potential, Mitochondrial/physiology , Mitochondrial Membranes/metabolism , Humans
19.
Proteomics ; 24(9): e2300312, 2024 May.
Article in English | MEDLINE | ID: mdl-38446070

ABSTRACT

The ectoparasitic mite Varroa destructor transmits and triggers viral infections that have deleterious effects on honey bee colonies worldwide. We performed a manipulative experiment in which worker bees collected at emergence were exposed to Varroa for 72 h, and their proteomes were compared with those of untreated control bees. Label-free quantitative proteomics identified 77 differentially expressed A. mellifera proteins (DEPs). In addition, viral proteins were identified by orthogonal analysis, and most importantly, Deformed wing virus (DWV) was found at high levels/intensity in Varroa-exposed bees. Pathway enrichment analysis suggested that the main pathways affected included peroxisomal metabolism, cyto-/exoskeleton reorganization, and cuticular proteins. Detailed examination of individual DEPs revealed that additional changes in DEPs were associated with peroxisomal function. In addition, the proteome data support the importance of TGF-ß signaling in Varroa-DWV interaction and the involvement of the mTORC1 and Hippo pathways. These results suggest that the effect of DWV on bees associated with Varroa feeding results in aberrant autophagy. In particular, autophagy is selectively modulated by peroxisomes, to which the observed proteome changes strongly corresponded. This study complements previous research with different study designs and suggests the importance of the peroxisome, which plays a key role in viral infections.


Subject(s)
Peroxisomes , RNA Viruses , Varroidae , Animals , Bees/virology , Bees/parasitology , Varroidae/virology , Peroxisomes/metabolism , Peroxisomes/virology , RNA Viruses/physiology , Proteomics/methods , Proteome/metabolism , Proteome/analysis , Insect Proteins/metabolism , Signal Transduction , Host-Parasite Interactions
20.
Methods Mol Biol ; 2776: 107-134, 2024.
Article in English | MEDLINE | ID: mdl-38502500

ABSTRACT

Plastids are organelles delineated by two envelopes playing important roles in different cellular processes such as energy production or lipid biosynthesis. To regulate their biogenesis and their function, plastids have to communicate with other cellular compartments. This communication can be mediated by metabolites, signaling molecules, and by the establishment of direct contacts between the plastid envelope and other organelles such as the endoplasmic reticulum, mitochondria, peroxisomes, plasma membrane, and the nucleus. These interactions are highly dynamic and respond to different biotic and abiotic stresses. However, the mechanisms involved in the formation of plastid-organelle contact sites and their functions are still far from being understood. In this chapter, we summarize our current knowledge about plastid contact sites and their role in the regulation of plastid biogenesis and function.


Subject(s)
Endoplasmic Reticulum , Plastids , Plastids/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Cell Membrane/metabolism , Peroxisomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...