1
|
Rattanasri A, Paensuwan P, Schamel WWWA, Pongcharoen S, Ngoenkam J. Nck1 Regulates Glucose Metabolism in Primary Human T Cells. Immunology 2025. [PMID: 40421785 DOI: 10.1111/imm.13950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 04/01/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
Non-catalytic region of tyrosine kinase 1 (Nck1) is an adaptor protein found in many cell types and plays several functions. In T cells, Nck1 is functionally associated with a T cell receptor (TCR)-mediated actin rearrangement, insulin signalling, PI3K/Akt/mTOR pathway, and lipid production. However, the role of Nck1 in regulating glucose metabolism in T cells is still largely unknown. In the present study, the role of Nck1 in glucose metabolism in primary human T cells was investigated. Plasmid encoding Nck1-specific short hairpin RNA (shRNA) was delivered to primary T cells to mediate Nck1 silencing. Plasmids encoding Nck1-specific short hairpin RNA (shRNA) were delivered to primary human T cells to mediate Nck1 silencing. Nck1-knockdown (N1KD) cells were analysed for processes related to glucose metabolism and function. Despite an increased expression of glucose transporter 1 (GLUT1) in N1KD cells, these cells exhibited impaired glucose uptake and ATP production, indicating dysfunction of GLUT1 or altered intracellular glucose metabolism. Nck1 depletion disrupted metabolic signalling characterised by reduced TXNIP and phosphoribosomal protein S6 (pS6) levels, along with an increased phosphorylation of Akt and AMPK. The reduced extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) found in N1KD cells indicated impaired glycolysis and oxidative phosphorylation. Functionally, these metabolic alterations were associated with impaired T cell activation, reduced proliferation, and increased apoptosis. Collectively, Nck1 critically regulated glucose metabolism in T cells, linking metabolic reprogramming to immune function and cell survival.
Collapse
Affiliation(s)
- Araya Rattanasri
- Graduate School of Biomedical Sciences Programme, Faculty of Allied Health Science, Naresuan University, Phitsanulok, Thailand
| | - Pussadee Paensuwan
- Department of Optometry, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Wolfgang W W A Schamel
- Signaling Research Centers BIOSS and CIBSS, Department of Immunology, Faculty of Biology, and Centre for Chronic Immunodeficiency (CCI), Freiburg University Clinics, University of Freiburg, Freiburg, Germany
| | - Sutatip Pongcharoen
- Division of Immunology, Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok, Thailand
| | - Jatuporn Ngoenkam
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
2
|
Nagashima Y, Sharma V, Reekers LF, von Schaewen A, Koiwa H. Kifunensine-sensitive ADP-ribosylation factor A1EG69R mutant reveals coordination of protein glycosylation and vesicle transport pathways. JOURNAL OF EXPERIMENTAL BOTANY 2025; 76:2112-2128. [PMID: 39807841 DOI: 10.1093/jxb/eraf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/11/2025] [Indexed: 01/16/2025]
Abstract
Complex N-glycans are asparagine (N)-linked branched sugar chains attached to secretory proteins in eukaryotes. They are produced by modification of N-linked oligosaccharide structures in the endoplasmic reticulum and Golgi apparatus. Complex N-glycans formed in the Golgi apparatus are often assigned specific roles unique to the host organism, with their roles in plants remaining largely unknown. Using inhibitor (kifunensine, KIF) hypersensitivity as read out, we identified Arabidopsis mutants that require complex N-glycan modification. Among >100 KIF-sensitive mutants, one showing abnormal secretory organelles and a salt-sensitive phenotype contained a point mutation leading to amino acid replacement (G69R) in ARFA1E, a small Arf1-GTPase family protein presumably involved in vesicular transport. In vitro assays showed that the G69R exchange interferes with protein activation. In vivo, ARFA1EG69R caused dominant-negative effects, altering the morphology of the endoplasmic reticulum, Golgi apparatus, and trans-Golgi network (TGN). Post-Golgi transport (endocytosis/endocytic recycling) of the essential glycoprotein KORRIGAN1, one of the KIF sensitivity targets, is slowed down constitutively as well as under salt stress in the ARFA1EG69R mutant. Because regulated cycling of plasma membrane proteins is required for stress tolerance of the host plants, the ARFA1EG69R mutant established a link between KIF-targeted luminal glycoprotein functions/dynamics and cytosolic regulators of vesicle transport in endosome-/cell wall-associated tolerance mechanisms.
Collapse
Affiliation(s)
- Yukihiro Nagashima
- Vegetable and Fruit Improvement Center and Department of Horticultural Sciences Texas A&M University, College Station, TX 77843, USA
| | - Vinita Sharma
- Vegetable and Fruit Improvement Center and Department of Horticultural Sciences Texas A&M University, College Station, TX 77843, USA
| | - Lea-Franziska Reekers
- Institut für Biologie und Biotechnologie der Pflanzen, Universität Münster, D-48149 Münster, Germany
| | - Antje von Schaewen
- Institut für Biologie und Biotechnologie der Pflanzen, Universität Münster, D-48149 Münster, Germany
| | - Hisashi Koiwa
- Vegetable and Fruit Improvement Center and Department of Horticultural Sciences Texas A&M University, College Station, TX 77843, USA
- Molecular and Environmental Plant Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
3
|
Pinheiro-Lustosa R, Gondim-Pereira NMS, Alves SADS, Takiya CM, da Silva-Alves KS, Pinheiro AAS, Coelho-de-Souza AN, Moreira-Gomes MD, Caruso-Neves C, Leal-Cardoso JH. Effects of Anethole on Renal Function of Swiss Mice. Pharmaceuticals (Basel) 2025; 18:541. [PMID: 40283976 PMCID: PMC12030660 DOI: 10.3390/ph18040541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Anethole, a terpenoid with several pharmacologic effects, is the major constituent of the essential oil of Croton zehntneri (EOCz), Pax & K. Hoffm, Euphorbiaceae. Due to the mild renal toxicity associated with high doses of EOCz, its potential therapeutic effects on several diseases, and the fact that its chemical composition consists of 80% anethole, the renal effects of anethole in mice were investigated. Methods: Mice were randomly divided into eight groups, dosed daily as follows: Group 1-CTRL (control; vehicle only); Groups 2-A100, 3-A1252x, and 4-A250 (dosed with 100, 125 twice daily, and 250 mg/kg, per os anethole); Group 5-SUBAKI (i.p. albumin to induce hyperproteinemia and proteinuria; subclinical acute kidney injury); and Groups 6-SUBAKI+A100, 7-SUBAKI+A1252x, and 8-SUBAKI+A250 (per os anethole + i.p. albumin). Results: The A1252x and A250 groups significantly increased urinary proteinuria and interstitial inflammation (p < 0.001, for these groups). SUBAKI+A100, SUBAKI+A1252x, and SUBAKI+A250 showed a neither protective nor additive effect in the proteinuria induced by anethole and by administered albumin. The anethole-induced proteinuria was spontaneously reversible in approximately 4 weeks. In vitro experiments showed that anethole (300 µg/mL) inhibits albumin uptake from the culture medium by tubular cells. Conclusions: Anethole at high doses bears renal acute toxicity that, although mild and spontaneously fully reversible, must be taken into consideration in a cost-benefit analysis.
Collapse
Affiliation(s)
- Romário Pinheiro-Lustosa
- Superior Institute of Biomedical Sciences, Universidade Estadual do Ceará, Fortaleza 60714-903, Brazil; (R.P.-L.); (N.M.S.G.-P.); (K.S.d.S.-A.); (A.N.C.-d.-S.); (M.D.M.-G.)
| | - Neide Maria Silva Gondim-Pereira
- Superior Institute of Biomedical Sciences, Universidade Estadual do Ceará, Fortaleza 60714-903, Brazil; (R.P.-L.); (N.M.S.G.-P.); (K.S.d.S.-A.); (A.N.C.-d.-S.); (M.D.M.-G.)
| | - Sarah Aparecida dos Santos Alves
- Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-617, Brazil; (S.A.d.S.A.); (C.M.T.); (A.A.S.P.); (C.C.-N.)
| | - Christina Maeda Takiya
- Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-617, Brazil; (S.A.d.S.A.); (C.M.T.); (A.A.S.P.); (C.C.-N.)
| | - Kerly Shamyra da Silva-Alves
- Superior Institute of Biomedical Sciences, Universidade Estadual do Ceará, Fortaleza 60714-903, Brazil; (R.P.-L.); (N.M.S.G.-P.); (K.S.d.S.-A.); (A.N.C.-d.-S.); (M.D.M.-G.)
| | - Ana Acacia Sá Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-617, Brazil; (S.A.d.S.A.); (C.M.T.); (A.A.S.P.); (C.C.-N.)
| | - Andrelina Noronha Coelho-de-Souza
- Superior Institute of Biomedical Sciences, Universidade Estadual do Ceará, Fortaleza 60714-903, Brazil; (R.P.-L.); (N.M.S.G.-P.); (K.S.d.S.-A.); (A.N.C.-d.-S.); (M.D.M.-G.)
| | - Maria Diana Moreira-Gomes
- Superior Institute of Biomedical Sciences, Universidade Estadual do Ceará, Fortaleza 60714-903, Brazil; (R.P.-L.); (N.M.S.G.-P.); (K.S.d.S.-A.); (A.N.C.-d.-S.); (M.D.M.-G.)
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-617, Brazil; (S.A.d.S.A.); (C.M.T.); (A.A.S.P.); (C.C.-N.)
| | - José Henrique Leal-Cardoso
- Superior Institute of Biomedical Sciences, Universidade Estadual do Ceará, Fortaleza 60714-903, Brazil; (R.P.-L.); (N.M.S.G.-P.); (K.S.d.S.-A.); (A.N.C.-d.-S.); (M.D.M.-G.)
| |
Collapse
|
4
|
Clouser PR, Riggs CL, Romney ALT, Podrabsky JE. Diapause and Anoxia-Induced Quiescence Are Unique States in Embryos of the Annual Killifish, Austrofundulus limnaeus. Biomolecules 2025; 15:515. [PMID: 40305273 PMCID: PMC12024583 DOI: 10.3390/biom15040515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/20/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025] Open
Abstract
Diapause is a state of developmental and metabolic dormancy that precedes exposure to environmental stresses. Yet, diapausing embryos are typically stress-tolerant. Evidence suggests that diapausing embryos "prepare" for stress as part of a gene expression program as they enter dormancy. Here, we investigate if diapause II embryos of the annual killifish Austrofundulus limnaeus, which can survive for hundreds of days of anoxia, can mount a transcriptomic response to anoxic insult. Bulk RNAseq was used to characterize the transcriptomes of diapause II embryos exposed to normoxia, 4 h and 24 h anoxia, and 2 h and 24 h normoxic recovery from anoxia. Differential expression and gene ontology analyses were used to probe for pathways that may mitigate survival. Transcriptional factor analysis was used to predict potential mediators of this response. Diapausing embryos exhibited a robust transcriptomic response to anoxia and recovery that returns to near baseline conditions after 24 h. Anoxia induced an upregulation of genes involved in the integrated stress response, lipid metabolism, p38mapk kinase signaling, and apoptosis. Developmental and mitochondrial genes decreased. We conclude that diapause II embryos mount a robust transcriptomic stress response when faced with anoxic insult. This response is consistent with mediating expected challenges to cellular homeostasis in anoxia.
Collapse
Affiliation(s)
- Patrick R. Clouser
- Department of Biology, Center for Life in Extreme Environments, Portland State University, P.O. Box 751, Portland, OR 97201, USA;
| | - Claire L. Riggs
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA 02115, USA;
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Amie L. T. Romney
- Department of Biology, Center for Life in Extreme Environments, Portland State University, P.O. Box 751, Portland, OR 97201, USA;
| | - Jason E. Podrabsky
- Department of Biology, Center for Life in Extreme Environments, Portland State University, P.O. Box 751, Portland, OR 97201, USA;
| |
Collapse
|
5
|
Milne SM, Edeen PT, Fay DS. TAT-1, a phosphatidylserine flippase, affects molting and regulates membrane trafficking in the epidermis of Caenorhabditis elegans. Genetics 2025; 229:iyae216. [PMID: 39722491 DOI: 10.1093/genetics/iyae216] [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: 09/15/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Membrane trafficking is a conserved process required for the import, export, movement, and distribution of proteins and other macromolecules within cells. The Caenorhabditis elegans NIMA-related kinases NEKL-2 (human NEK8/9) and NEKL-3 (human NEK6/7) are conserved regulators of membrane trafficking and are required for the completion of molting. Using a genetic approach, we identified reduction-of-function mutations in tat-1 that suppress nekl-associated molting defects. tat-1 encodes the C. elegans ortholog of mammalian ATP8A1/2, a phosphatidylserine flippase that promotes the asymmetric distribution of phosphatidylserine on the cytosolic leaflet of lipid membrane bilayers. CHAT-1 (human CDC50), a conserved chaperone, was required for the correct localization of TAT-1, and chat-1 inhibition strongly suppressed nekl defects. Using a phosphatidylserine sensor, we found that TAT-1 was required for the normal localization of phosphatidylserine at apical endosomes and that loss of TAT-1 led to aberrant endosomal morphologies. Consistent with these data, TAT-1 localized to early endosomes and to recycling endosomes marked with RME-1, the C. elegans ortholog of the human EPS15 homology domain-containing protein, EHD1. TAT-1, phosphatidylserine biosynthesis, and the phosphatidylserine-binding protein RFIP-2 (human RAB11-FIP2) were all required for the normal localization of RME-1 to apical endosomes. Consistent with these proteins functioning together, inhibition of RFIP-2 or RME-1 led to the partial suppression of nekl molting defects, as did inhibition of phosphatidylserine biosynthesis. We propose that TAT-1 flippase activity, in conjunction with RFIP-2, promotes the recruitment of RME-1 to apical recycling endosomes and that inhibition of TAT-1-RFIP-2-RME-1 can compensate for a reduction in NEKL activities.
Collapse
Affiliation(s)
- Shae M Milne
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| | - Philip T Edeen
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| | - David S Fay
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| |
Collapse
|
6
|
Sao K, Risbud MV. SDC4 drives fibrotic remodeling of the intervertebral disc under altered spinal loading. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643128. [PMID: 40161806 PMCID: PMC11952502 DOI: 10.1101/2025.03.13.643128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Alterations in physiological loading of the spine are deleterious to intervertebral disc health. The caudal spine region Ca3-6 that experiences increased flexion, showed disc degeneration in young adult mice. Given the role of Syndecan 4 (SDC4), a cell surface heparan sulfate proteoglycan in disc matrix catabolism and mechanosensing, we investigated if deletion could mitigate this loading-dependent phenotype. Notably, at spinal levels Ca3-6, Sdc4- KO mice did not exhibit increased collagen fibril and fibronectin deposition in the NP compartment or showed the alterations in collagen crosslinks observed in wild-type mice. Similarly, unlike wild-type mice, NP cells in Sdc4 -KO mice retained transgelin (TGLN) expression and showed absence of COL X deposition, pointing to the preservation of their notochordal characteristics. Proteomic analysis revealed that NP tissues responded to the abnormal loading by increasing the abundance of proteins associated with extracellular matrix remodeling, chondrocyte development, and contractility. Similarly, downregulated proteins suggested decreased vesicle transport, autophagy-related pathway, and RNA quality control regulation. Notably, NP proteome from Sdc4 KO suggested that increased dynamin-mediated endocytosis, autophagy-related pathway, and RNA and DNA quality control may underscore the protection from increased flexion-induced degeneration. Our study highlights the important role of SDC4 in fine-tuning cellular homeostasis and extracellular matrix production in disc environment subjected to altered loading.
Collapse
|
7
|
Ruzzante B, Fruzzetti F, Cattaneo M, Lauria Pinter G, Marcuzzo S, Candiani G, Bono N. Harnessing osmotic shock for enhanced intracellular delivery of (nano)cargos. Int J Pharm 2025; 669:125008. [PMID: 39638270 DOI: 10.1016/j.ijpharm.2024.125008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Efficient intracellular delivery of exogenous (nano)materials is critical for both research and therapeutic applications. The physicochemical properties of the cargo play a crucial role in determining internalization efficacy. Consequently, significant research efforts are focused on developing innovative and effective methodologies to optimize (nano)material delivery. In this study, we utilized osmotic shock to enhance (nano)cargos internalization. We examined the effects of hypotonic/hypertonic shock on both primary and cell lines, assessing parameters such as cell viability, cell volume, membrane tension changes, and particle uptake. Our results indicate that short-lived osmotic shock does not harm cells. Hypotonic shock induced temporary shape changes lasting up to 5 min, followed by a 15-minute recovery period. Importantly, hypotonic shock increased the uptake of 100-nm and 500-nm particles by ∼ 3- and ∼ 5-fold, respectively, compared to isotonic conditions. In contrast, the hypertonic shock did not impact cell behavior or particle uptake. Notably, the internalization mechanisms triggered by osmotic shock operate independently of active endocytic pathways, making hypotonic stimulation particularly beneficial for hard-to-treat cells. When primary fibroblasts derived from amyotrophic lateral sclerosis (ALS)-patients were exposed to hypotonic shock in the presence of the therapeutic cargo icerguastat, there was an increased expression of miR-106b-5p compared to isotonic conditions. In conclusion, osmotic shock presents a promising strategy for improving drug delivery within cells and, potentially, in tissues such as muscles or skin, where localized drug administration is preferred.
Collapse
Affiliation(s)
- Beatrice Ruzzante
- genT_LΛB, Dept. of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Flaminia Fruzzetti
- genT_LΛB, Dept. of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Marco Cattaneo
- ALS Centre, 3rd Neurology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy; Ph.D. Program in Pharmacological Biomolecular Sciences, Experimental and Clinical, University of Milan, Milan, Italy
| | - Giuseppe Lauria Pinter
- ALS Centre, 3rd Neurology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Stefania Marcuzzo
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy; Brain-targeted Nanotechnologies (BraiNs) Lab, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Gabriele Candiani
- genT_LΛB, Dept. of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy; Brain-targeted Nanotechnologies (BraiNs) Lab, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Nina Bono
- genT_LΛB, Dept. of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy.
| |
Collapse
|
8
|
Chauhan H, Carruthers NJ, Stemmer PM, Schneider BL, Moszczynska A. Interactions of VMAT2 with CDCrel-1 and Parkin in Methamphetamine Neurotoxicity. Int J Mol Sci 2024; 25:13070. [PMID: 39684782 DOI: 10.3390/ijms252313070] [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: 08/25/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 12/18/2024] Open
Abstract
In recent years, methamphetamine (METH) misuse in the US has been rapidly increasing, and there is no FDA-approved pharmacotherapy for METH use disorder (MUD). In addition to being dependent on the drug, people with MUD develop a variety of neurological problems related to the toxicity of this drug. A variety of molecular mechanisms underlying METH neurotoxicity has been identified, including the dysfunction of the neuroprotective protein parkin. However, it is not known whether parkin loss of function within striatal dopaminergic (DAergic) terminals translates into decreased DA storage capacity. This study examined the relationship between parkin, its substrate cell division cycle related-1 (CDCrel-1) associated with synaptic vesicles, and vesicular monoamine transporter-2 (VMAT2) responsible for packaging DA in an in vivo model of METH neurotoxicity. To assess the individual differences in response to METH's neurotoxic effects, a large group of male Sprague Dawley rats were treated with binge METH or saline and sacrificed 1 h or 24 h later. This study is the first to show that CDCrel-1 interacts with VMAT2 in the rat striatum and that binge METH can alter this interaction as well as the levels and subcellular localization of CDCrel-1. The proteomic analysis of VMAT-2-associated proteins revealed the upregulation of several proteins involved in the exocytosis/endocytosis cycle and responses to stress. The results suggest that DAergic neurons are engaged in counteracting METH-induced toxic effects, including attempts to increase endocytosis and autophagy at 1 h after the METH binge, with the responses varying widely between individual rats. Studying CDCrel-1, VMAT2, and other proteins in large groups of outbred rats can help define individual genetic and molecular differences in responses to METH neurotoxicity, which, in turn, may aid treating humans suffering from MUD and its neurological consequences.
Collapse
Affiliation(s)
- Heli Chauhan
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave., Detroit, MI 48201, USA
| | - Nicholas J Carruthers
- Institute of Environmental Health Sciences and Proteomics Core Facility, 540 East Canfield Ave., Detroit, MI 48202, USA
- Bioinformatics Core, Michigan Medicine, University of Michigan, NCRC Building 14, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA
| | - Paul M Stemmer
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave., Detroit, MI 48201, USA
- Institute of Environmental Health Sciences and Proteomics Core Facility, 540 East Canfield Ave., Detroit, MI 48202, USA
| | - Bernard L Schneider
- Bertarelli Platform for Gene Therapy, École Polytechnique Fédérale de Lausanne, School of Life Sciences, Ch. Des Mines 9, CH-1202 Geneva, Switzerland
| | - Anna Moszczynska
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave., Detroit, MI 48201, USA
| |
Collapse
|
9
|
Damoo D, Kretschmer M, Lee CWJ, Herrfurth C, Feussner I, Heimel K, Kronstad JW. Herbicides as fungicides: Targeting heme biosynthesis in the maize pathogen Ustilago maydis. MOLECULAR PLANT PATHOLOGY 2024; 25:e70007. [PMID: 39487654 PMCID: PMC11530707 DOI: 10.1111/mpp.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 11/04/2024]
Abstract
Pathogens must efficiently acquire nutrients from host tissue to proliferate, and strategies to block pathogen access therefore hold promise for disease control. In this study, we investigated whether heme biosynthesis is an effective target for ablating the virulence of the phytopathogenic fungus Ustilago maydis on maize plants. We first constructed conditional heme auxotrophs of the fungus by placing the heme biosynthesis gene hem12 encoding uroporphyrinogen decarboxylase (Urod) under the control of nitrogen or carbon source-regulated promoters. These strains were heme auxotrophs under non-permissive conditions and unable to cause disease in maize seedlings, thus demonstrating the inability of the fungus to acquire sufficient heme from host tissue to support proliferation. Subsequent experiments characterized the role of endocytosis in heme uptake, the susceptibility of the fungus to heme toxicity as well as the transcriptional response to exogenous heme. The latter RNA-seq experiments identified a candidate ABC transporter with a role in the response to heme and xenobiotics. Given the importance of heme biosynthesis for U. maydis pathogenesis, we tested the ability of the well-characterized herbicide BroadStar to influence disease. This herbicide contains the active ingredient flumioxazin, an inhibitor of Hem14 in the heme biosynthesis pathway, and we found that it was an effective antifungal agent for blocking disease in maize. Thus, repurposing herbicides for which resistant plants are available may be an effective strategy to control pathogens and achieve crop protection.
Collapse
Affiliation(s)
- Djihane Damoo
- Michael Smith Laboratories, Department of Microbiology and ImmunologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Matthias Kretschmer
- Michael Smith Laboratories, Department of Microbiology and ImmunologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Christopher W. J. Lee
- Michael Smith Laboratories, Department of Microbiology and ImmunologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Cornelia Herrfurth
- Department of Plant Biochemistry, Albrecht‐von‐Haller Institute of Plant SciencesUniversity of GöttingenGöttingenGermany
- Service Unit for Metabolomics and Lipidomics, Göttingen Center for Molecular Biosciences (GZMB)University of GöttingenGöttingenGermany
| | - Ivo Feussner
- Department of Plant Biochemistry, Albrecht‐von‐Haller Institute of Plant SciencesUniversity of GöttingenGöttingenGermany
- Service Unit for Metabolomics and Lipidomics, Göttingen Center for Molecular Biosciences (GZMB)University of GöttingenGöttingenGermany
- Department of Plant Biochemistry, Göttingen Center for Molecular Biosciences (GZMB)University of GöttingenGöttingenGermany
| | - Kai Heimel
- Institute of Microbiology and Genetics, Department of Microbial Cell Biology, Göttingen Center for Molecular Biosciences (GZMB)University of GöttingenGöttingenGermany
| | - James W. Kronstad
- Michael Smith Laboratories, Department of Microbiology and ImmunologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
10
|
Milne SM, Edeen PT, Fay DS. TAT-1, a phosphatidylserine flippase, affects molting and regulates membrane trafficking in the epidermis of C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613099. [PMID: 39314363 PMCID: PMC11419146 DOI: 10.1101/2024.09.15.613099] [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
Membrane trafficking is a conserved process required for the movement and distribution of proteins and other macromolecules within cells. The Caenorhabditis elegans NIMA-related kinases NEKL-2 (human NEK8/9) and NEKL-3 (human NEK6/7) are conserved regulators of membrane trafficking and are required for the completion of molting. We used a genetic approach to identify reduction-of-function mutations in tat-1 that suppress nekl -associated molting defects. tat-1 encodes the C. elegans ortholog of mammalian ATP8A1/2, a phosphatidylserine (PS) flippase that promotes the asymmetric distribution of PS to the cytosolic leaflet of lipid membrane bilayers. CHAT-1 (human CDC50), a conserved chaperone, was required for the correct localization of TAT-1, and chat-1 inhibition strongly suppressed nekl defects. Using a PS sensor, we found that TAT-1 was required for the normal localization of PS at apical endosomes and that loss of TAT-1 led to aberrant endosomal morphologies. Consistent with this, TAT-1 localized to early endosomes and to recycling endosomes marked with RME-1, the C. elegans ortholog of the human EPS15 homology (EH) domain-containing protein, EHD1. TAT-1, PS biosynthesis, and the PS-binding protein RFIP-2 (human RAB11-FIP2) were all required for the normal localization of RME-1 to apical endosomes. Consistent with these proteins functioning together, inhibition of RFIP-2 or RME-1 led to the partial suppression of nekl molting defects, as did the inhibition of PS biosynthesis. Using the auxin-inducible degron system, we found that depletion of NEKL-2 or NEKL-3 led to defects in RME-1 localization and that a reduction in TAT-1 function partially restored RME-1 localization in NEKL-3-depleted cells. ARTICLE SUMMARY Endocytosis is an essential process required for the movement of proteins and lipids within cells. NEKL-2 and NEKL-3, two evolutionarily conserved proteins in the nematode Caenorhabditis elegans , are important regulators of endocytosis. In the current study, the authors describe a new functional link between the NEKLs and several proteins with known roles in endocytosis including TAT-1, a conserved enzyme that moves lipids between the bilayers of cellular membranes. As previous work implicated NEKLs in developmental defects and cancer, the present study can provide new insights into how the misregulation of endocytosis affects human health and disease.
Collapse
|
11
|
van Belle GJ, Zieseniss A, Heidenreich D, Olmos M, Zhuikova A, Möbius W, Paul MW, Katschinski DM. Cargo-specific effects of hypoxia on clathrin-mediated trafficking. Pflugers Arch 2024; 476:1399-1410. [PMID: 38294517 PMCID: PMC11310247 DOI: 10.1007/s00424-024-02911-6] [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: 07/04/2023] [Revised: 12/18/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024]
Abstract
Clathrin-associated trafficking is a major mechanism for intracellular communication, as well as for cells to communicate with the extracellular environment. A decreased oxygen availability termed hypoxia has been described to influence this mechanism in the past. Mostly biochemical studies were applied in these analyses, which miss spatiotemporal information. We have applied live cell microscopy and a newly developed analysis script in combination with a GFP-tagged clathrin-expressing cell line to obtain insight into the dynamics of the effect of hypoxia. Number, mobility and directionality of clathrin-coated vesicles were analysed in non-stimulated cells as well as after stimulation with epidermal growth factor (EGF) or transferrin in normoxic and hypoxic conditions. These data reveal cargo-specific effects, which would not be observable with biochemical methods or with fixed cells and add to the understanding of cell physiology in hypoxia. The stimulus-dependent consequences were also reflected in the final cellular output, i.e. decreased EGF signaling and in contrast increased iron uptake in hypoxia.
Collapse
Affiliation(s)
- Gijsbert J van Belle
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August University, 37073, Göttingen, Germany
| | - Anke Zieseniss
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August University, 37073, Göttingen, Germany
| | - Doris Heidenreich
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August University, 37073, Göttingen, Germany
| | - Maxime Olmos
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August University, 37073, Göttingen, Germany
| | - Asia Zhuikova
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August University, 37073, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Electron Microscopy, City Campus, Max-Planck-Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Maarten W Paul
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dörthe M Katschinski
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August University, 37073, Göttingen, Germany.
| |
Collapse
|
12
|
Surati U, Niranjan SK, Pundir RK, Koul Y, Vohra V, Gandham RK, Kumar A. Genome-wide comparative analyses highlight selection signatures underlying saline adaptation in Chilika buffalo. Physiol Genomics 2024; 56:609-620. [PMID: 38949516 DOI: 10.1152/physiolgenomics.00028.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/30/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024] Open
Abstract
Chilika, a native buffalo breed of the Eastern coast of India, is mainly distributed around the Chilika brackish water lake connected with the Bay of Bengal Sea. This breed possesses a unique ability to delve deep into the salty water of the lake and stay there to feed on local vegetation of saline nature. Adaptation to salinity is a genetic phenomenon; however, the genetic basis underlying salinity tolerance is still limited in animals, specifically in livestock. The present study explores the genetic evolution that unveils the Chilika buffalo's adaptation to the harsh saline habitat, including both water and food systems. For this study, whole genome resequencing data on 18 Chilika buffalo and for comparison 10 Murrah buffalo of normal habitat were generated. For identification of selection sweeps, intrapopulation and interpopulation statistics were used. A total of 709, 309, 468, and 354 genes were detected to possess selection sweeps in Chilika buffalo using the nucleotide diversity (θπ), Tajima's D, nucleotide diversity ratio (θπ-ratio), and FST methods, respectively. Further analysis revealed a total of 23 genes including EXOC6B, VPS8, LYPD1, VPS35, CAMKMT, NCKAP5, COMMD1, myosin light chain kinase 3 (MYLK3), and B3GNT2 were found to be common by all the methods. Furthermore, functional annotation study of identified genes provided pathways such as MAPK signaling, renin secretion, endocytosis, oxytocin signaling pathway, etc. Gene network analysis enlists that hub genes provide insights into their interactions with each other. In conclusion, this study has highlighted the genetic basis underlying the local adaptive function of Chilika buffalo under saline environment.NEW & NOTEWORTHY Indian Chilika buffaloes are being maintained on extensive grazing system and have a unique ability to convert local salty vegetation into valuable human food. However, adaptability to saline habitat of Chilika buffalo has not been explored to date. Here, we identified genes and biological pathways involved, such as MAPK signaling, renin secretion, endocytosis, and oxytocin signaling pathway, underlying adaptability of Chilika buffalo to saline environment. This investigation shed light on the mechanisms underlying the buffalo's resilience in its native surroundings.
Collapse
Affiliation(s)
- Utsav Surati
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
- ICAR-National Dairy Research Institute, Karnal, India
| | | | | | - Ymberzal Koul
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
- ICAR-National Dairy Research Institute, Karnal, India
| | - Vikas Vohra
- ICAR-National Dairy Research Institute, Karnal, India
| | | | - Amod Kumar
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| |
Collapse
|
13
|
Chauhan H, Carruthers N, Stemmer P, Schneider BP, Moszczynska A. Neurotoxic Methamphetamine Doses Alter CDCel-1 Levels and Its Interaction with Vesicular Monoamine Transporter-2 in Rat Striatum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.21.604458. [PMID: 39091864 PMCID: PMC11291068 DOI: 10.1101/2024.07.21.604458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
In recent years, methamphetamine METH misuse in the US has been rapidly increasing and there is no FDA-approved pharmacotherapy for METH use disorder (MUD). In addition to being dependent on the drug, people with MUD develop a variety of neurological problems related to the toxicity of this drug. A variety of molecular mechanisms underlying METH neurotoxicity has been identified, including dysfunction of the neuroprotective protein parkin. However, it is not known whether parkin loss of function within striatal dopaminergic (DAergic) terminals translates into a decrease in DA storage capacity. This study examined the relationship between parkin, its substrate cell division cycle related-1 (CDCrel-1), and vesicular monoamine transporter-2 (VMAT2) in METH neurotoxicity in male Sprague Dawley rats. To also assess individual differences in response to METH's neurotoxic effects, a large group of rats was treated with binge METH or saline and sacrificed 1h or 24h later. This study is the first to show that binge METH alters the levels and subcellular localization of CDCrel-1 and that CDCrel-1 interacts with VMAT2 and increases its levels at the plasma membrane. Furthermore, we found wide individual differences in the responses of measured indices to METH. Proteomic analysis of VMAT-2-associated proteins revealed upregulation of several proteins involved in the exocytosis/endocytosis cycle. The results suggest that at 1h after METH binge, DAergic neurons are engaged in counteracting METH-induced toxic effects, including oxidative stress- and hyperthermia-induced inhibition of synaptic vesicle cycling, with the responses varying between individual rats. Studying CDCrel-1, VMAT2, and other proteins in large groups of outbred rats can help define individual genetic and molecular differences in responses to METH neurotoxicity which, in turn, will aid treating humans suffering from METH use disorder and its neurological consequences.
Collapse
Affiliation(s)
- Heli Chauhan
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI, USA 48201
| | - Nick Carruthers
- Institute of Environmental Health Sciences and Proteomics Core Facility, 540 East Canfield Ave., Detroit, MI 48202
| | - Paul Stemmer
- Institute of Environmental Health Sciences and Proteomics Core Facility, 540 East Canfield Ave., Detroit, MI 48202
| | - Bernard P. Schneider
- Brain Mind Institute École Polytechnique Fédérale de Lausanne School of Life Sciences, Ch. Des Mines, 9, CH-1202 Geneve, Switzerland
| | - Anna Moszczynska
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI, USA 48201
| |
Collapse
|
14
|
Beaudreau N, Page TM, Drolet D, McKindsey CW, Howland KL, Calosi P. Using a metabolomics approach to investigate the sensitivity of a potential Arctic-invader and its Arctic sister-species to marine heatwaves and traditional harvesting disturbances. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 917:170167. [PMID: 38242480 DOI: 10.1016/j.scitotenv.2024.170167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/21/2023] [Accepted: 01/12/2024] [Indexed: 01/21/2024]
Abstract
Coastal species are threatened by fishing practices and changing environmental conditions, such as marine heatwaves (MHW). The mechanisms that confer tolerance to such stressors in marine invertebrates are poorly understood. However, differences in tolerance among different species may be attributed to their geographical distribution. To test the tolerance of species occupying different thermal ranges, we used two closely related bivalves the softshell clam Mya arenaria (Linnaeus, 1758), a cold-temperate invader with demonstrated potential for establishment in the Arctic, and the blunt gaper Mya truncata (Linnaeus, 1758), a native polar species. Clams were subjected to a thermal stress, mimicking a MHW, and harvesting stress in a controlled environment. Seven acute temperature changes (2, 7, 12, 17, 22, 27, and 32 °C) were tested at two harvesting disturbance intensities (with, without). Survival was measured after 12 days and three tissues (gills, mantle, and posterior adductor muscle) collected from surviving individuals for targeted metabolomic profiling. MHW tolerance differed significantly between species: 26.9 °C for M. arenaria and 17.8 °C for M. truncata, with a negligeable effect of harvesting. At the upper thermal limit, M. arenaria displayed a more profound metabolomic remodelling when compared to M. truncata, and this varied greatly between tissue types. Network analysis revealed differences in pathway utilization at the upper MHW limit, with M. arenaria displaying a greater reliance on multiple DNA repair and expression and cell signalling pathways, while M. truncata was limited to fewer pathways. This suggests that M. truncata is ill equipped to cope with warming environments. MHW patterning in the Northwest Atlantic may be a strong predictor of population survival and future range shifts in these two clam species. As polar environments undergo faster rates of warming compared to the global average, M. truncata may be outcompeted by M. arenaria expanding into its native range.
Collapse
Affiliation(s)
- Nicholas Beaudreau
- Laboratoire de Physiologie Écologique et Évolutive Marine, Département de Biologie, Chimie et Géographie, Université du Québec à Rimouski, Rimouski, Québec, Canada
| | - Tessa M Page
- Laboratoire de Physiologie Écologique et Évolutive Marine, Département de Biologie, Chimie et Géographie, Université du Québec à Rimouski, Rimouski, Québec, Canada
| | - David Drolet
- Fisheries and Oceans Canada, Demersal and Benthic Science Branch, Institut Maurice-Lamontagne, Mont-Joli, Québec, Canada
| | - Christopher W McKindsey
- Fisheries and Oceans Canada, Demersal and Benthic Science Branch, Institut Maurice-Lamontagne, Mont-Joli, Québec, Canada
| | - Kimberly L Howland
- Fisheries and Oceans Canada, Arctic and Aquatic Research Division, Freshwater Institute, Winnipeg, Manitoba, Canada
| | - Piero Calosi
- Laboratoire de Physiologie Écologique et Évolutive Marine, Département de Biologie, Chimie et Géographie, Université du Québec à Rimouski, Rimouski, Québec, Canada.
| |
Collapse
|
15
|
Keita VM, Lee YQ, Lakshmanan M, Ow DSW, Staniland P, Staniland J, Savill I, Tee KL, Wong TS, Lee DY. Evaluating oleaginous yeasts for enhanced microbial lipid production using sweetwater as a sustainable feedstock. Microb Cell Fact 2024; 23:63. [PMID: 38402186 PMCID: PMC10893622 DOI: 10.1186/s12934-024-02336-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/14/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Yeasts exhibit promising potential for the microbial conversion of crude glycerol, owing to their versatility in delivering a wide range of value-added products, particularly lipids. Sweetwater, a methanol-free by-product of the fat splitting process, has emerged as a promising alternative feedstock for the microbial utilization of crude glycerol. To further optimize sweetwater utilization, we compared the growth and lipid production capabilities of 21 oleaginous yeast strains under different conditions with various glycerol concentrations, sweetwater types and pH. RESULTS We found that nutrient limitation and the unique carbon composition of sweetwater boosted significant lipid accumulation in several strains, in particular Rhodosporidium toruloides NRRL Y-6987. Subsequently, to decipher the underlying mechanism, the transcriptomic changes of R. toruloides NRRL Y-6987 were further analyzed, indicating potential sugars and oligopeptides in sweetwater supporting growth and lipid accumulation as well as exogenous fatty acid uptake leading to the enhanced lipid accumulation. CONCLUSION Our comparative study successfully demonstrated sweetwater as a cost-effective feedstock while identifying R. toluroides NRRL Y-6987 as a highly promising microbial oil producer. Furthermore, we also suggested potential sweetwater type and strain engineering targets that could potentially enhance microbial lipid production.
Collapse
Affiliation(s)
- Valériane Malika Keita
- Department of Chemical & Biological Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, S1 3JD, UK
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros, Singapore, 138668, Singapore
| | - Yi Qing Lee
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros, Singapore, 138668, Singapore
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Dave Siak-Wei Ow
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros, Singapore, 138668, Singapore
| | - Paul Staniland
- Croda Europe Ltd., Oak Road, Clough Road, Hull, HU6 7PH, UK
| | | | - Ian Savill
- Croda Europe Ltd., Oak Road, Clough Road, Hull, HU6 7PH, UK
| | - Kang Lan Tee
- Department of Chemical & Biological Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, S1 3JD, UK
| | - Tuck Seng Wong
- Department of Chemical & Biological Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, S1 3JD, UK.
- Evolutor Ltd, The Innovation Centre, 217 Portobello, Sheffield, S1 4DP, UK.
- National Center for Genetic Engineering and Biotechnology, 113 Thailand Science Park, Phahonyothin Road, Khlong Nueang, Khlong Luang, 12120, Pathum Thani, Thailand.
- School of Pharmacy, Bandung Institute of Technology, 10 Coblong, Bandung, West Java, 40132, Indonesia.
| | - Dong-Yup Lee
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
| |
Collapse
|
16
|
Wenzel EM, Pedersen NM, Elfmark LA, Wang L, Kjos I, Stang E, Malerød L, Brech A, Stenmark H, Raiborg C. Intercellular transfer of cancer cell invasiveness via endosome-mediated protease shedding. Nat Commun 2024; 15:1277. [PMID: 38341434 PMCID: PMC10858897 DOI: 10.1038/s41467-024-45558-8] [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: 09/04/2022] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Overexpression of the transmembrane matrix metalloproteinase MT1-MMP/MMP14 promotes cancer cell invasion. Here we show that MT1-MMP-positive cancer cells turn MT1-MMP-negative cells invasive by transferring a soluble catalytic ectodomain of MT1-MMP. Surprisingly, this effect depends on the presence of TKS4 and TKS5 in the donor cell, adaptor proteins previously implicated in invadopodia formation. In endosomes of the donor cell, TKS4/5 promote ADAM-mediated cleavage of MT1-MMP by bridging the two proteases, and cleavage is stimulated by the low intraluminal pH of endosomes. The bridging depends on the PX domains of TKS4/5, which coincidently interact with the cytosolic tail of MT1-MMP and endosomal phosphatidylinositol 3-phosphate. MT1-MMP recruits TKS4/5 into multivesicular endosomes for their subsequent co-secretion in extracellular vesicles, together with the enzymatically active ectodomain. The shed ectodomain converts non-invasive recipient cells into an invasive phenotype. Thus, TKS4/5 promote intercellular transfer of cancer cell invasiveness by facilitating ADAM-mediated shedding of MT1-MMP in acidic endosomes.
Collapse
Affiliation(s)
- Eva Maria Wenzel
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Nina Marie Pedersen
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Liv Anker Elfmark
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ling Wang
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ingrid Kjos
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Espen Stang
- Laboratory for Molecular and Cellular Cancer Research, Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Lene Malerød
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Andreas Brech
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Section for Physiology and Cell Biology, Dept. of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Harald Stenmark
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Camilla Raiborg
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway.
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
17
|
Samakkarn W, Vandecruys P, Moreno MRF, Thevelein J, Ratanakhanokchai K, Soontorngun N. New biomarkers underlying acetic acid tolerance in the probiotic yeast Saccharomyces cerevisiae var. boulardii. Appl Microbiol Biotechnol 2024; 108:153. [PMID: 38240846 PMCID: PMC10799125 DOI: 10.1007/s00253-023-12946-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 01/22/2024]
Abstract
Evolutionary engineering experiments, in combination with omics technologies, revealed genetic markers underpinning the molecular mechanisms behind acetic acid stress tolerance in the probiotic yeast Saccharomyces cerevisiae var. boulardii. Here, compared to the ancestral Ent strain, evolved yeast strains could quickly adapt to high acetic acid levels (7 g/L) and displayed a shorter lag phase of growth. Bioinformatic-aided whole-genome sequencing identified genetic changes associated with enhanced strain robustness to acetic acid: a duplicated sequence in the essential endocytotic PAN1 gene, mutations in a cell wall mannoprotein (dan4Thr192del), a lipid and fatty acid transcription factor (oaf1Ser57Pro) and a thiamine biosynthetic enzyme (thi13Thr332Ala). Induction of PAN1 and its associated endocytic complex SLA1 and END3 genes was observed following acetic acid treatment in the evolved-resistant strain when compared to the ancestral strain. Genome-wide transcriptomic analysis of the evolved Ent acid-resistant strain (Ent ev16) also revealed a dramatic rewiring of gene expression among genes associated with cellular transport, metabolism, oxidative stress response, biosynthesis/organization of the cell wall, and cell membrane. Some evolved strains also displayed better growth at high acetic acid concentrations and exhibited adaptive metabolic profiles with altered levels of secreted ethanol (4.0-6.4% decrease), glycerol (31.4-78.5% increase), and acetic acid (53.0-60.3% increase) when compared to the ancestral strain. Overall, duplication/mutations and transcriptional alterations are key mechanisms driving improved acetic acid tolerance in probiotic strains. We successfully used adaptive evolutionary engineering to rapidly and effectively elucidate the molecular mechanisms behind important industrial traits to obtain robust probiotic yeast strains for myriad biotechnological applications. KEY POINTS: •Acetic acid adaptation of evolutionary engineered robust probiotic yeast S. boulardii •Enterol ev16 with altered genetic and transcriptomic profiles survives in up to 7 g/L acetic acid •Improved acetic acid tolerance of S. boulardii ev16 with mutated PAN1, DAN4, OAF1, and THI13 genes.
Collapse
Affiliation(s)
- Wiwan Samakkarn
- Excellent Research Laboratory for Yeast Innovation, Division of Biochemical Technology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Paul Vandecruys
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Heverlee, Belgium
- Center for Microbiology, VIB, Leuven, Flanders, Belgium
| | - Maria Remedios Foulquié Moreno
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Heverlee, Belgium
- Center for Microbiology, VIB, Leuven, Flanders, Belgium
| | - Johan Thevelein
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Heverlee, Belgium
- Center for Microbiology, VIB, Leuven, Flanders, Belgium
- NovelYeast Bv, Open Bio-Incubator, Erasmus High School, (Jette), Brussels, Belgium
| | - Khanok Ratanakhanokchai
- Excellent Research Laboratory for Yeast Innovation, Division of Biochemical Technology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
- Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Nitnipa Soontorngun
- Excellent Research Laboratory for Yeast Innovation, Division of Biochemical Technology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok, Thailand.
| |
Collapse
|
18
|
Liu MCJ, Yeh FLJ, Yvon R, Simpson K, Jordan S, Chambers J, Wu HM, Cheung AY. Extracellular pectin-RALF phase separation mediates FERONIA global signaling function. Cell 2024; 187:312-330.e22. [PMID: 38157854 DOI: 10.1016/j.cell.2023.11.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/01/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024]
Abstract
The FERONIA (FER)-LLG1 co-receptor and its peptide ligand RALF regulate myriad processes for plant growth and survival. Focusing on signal-induced cell surface responses, we discovered that intrinsically disordered RALF triggers clustering and endocytosis of its cognate receptors and FER- and LLG1-dependent endocytosis of non-cognate regulators of diverse processes, thus capable of broadly impacting downstream responses. RALF, however, remains extracellular. We demonstrate that RALF binds the cell wall polysaccharide pectin. They phase separate and recruit FER and LLG1 into pectin-RALF-FER-LLG1 condensates to initiate RALF-triggered cell surface responses. We show further that two frequently encountered environmental challenges, elevated salt and temperature, trigger RALF-pectin phase separation, promiscuous receptor clustering and massive endocytosis, and that this process is crucial for recovery from stress-induced growth attenuation. Our results support that RALF-pectin phase separation mediates an exoskeletal mechanism to broadly activate FER-LLG1-dependent cell surface responses to mediate the global role of FER in plant growth and survival.
Collapse
Affiliation(s)
- Ming-Che James Liu
- Department of Biochemistry and Molecular Biology, University of Massachusetts, 710 N. Pleasant St., Lederle Graduate Tower, Amherst, MA 01003, USA
| | - Fang-Ling Jessica Yeh
- Department of Biochemistry and Molecular Biology, University of Massachusetts, 710 N. Pleasant St., Lederle Graduate Tower, Amherst, MA 01003, USA; Plant Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Robert Yvon
- Department of Biochemistry and Molecular Biology, University of Massachusetts, 710 N. Pleasant St., Lederle Graduate Tower, Amherst, MA 01003, USA; Molecular and Cell Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Kelly Simpson
- Department of Biochemistry and Molecular Biology, University of Massachusetts, 710 N. Pleasant St., Lederle Graduate Tower, Amherst, MA 01003, USA; Molecular and Cell Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Samuel Jordan
- Department of Biochemistry and Molecular Biology, University of Massachusetts, 710 N. Pleasant St., Lederle Graduate Tower, Amherst, MA 01003, USA
| | - James Chambers
- Light Microscopy Core Facility, Institute of Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Hen-Ming Wu
- Department of Biochemistry and Molecular Biology, University of Massachusetts, 710 N. Pleasant St., Lederle Graduate Tower, Amherst, MA 01003, USA; Molecular and Cell Biology Program, University of Massachusetts, Amherst, MA 01003, USA.
| | - Alice Y Cheung
- Department of Biochemistry and Molecular Biology, University of Massachusetts, 710 N. Pleasant St., Lederle Graduate Tower, Amherst, MA 01003, USA; Molecular and Cell Biology Program, University of Massachusetts, Amherst, MA 01003, USA; Plant Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
19
|
Diaz J, Pellois JP. Deciphering variations in the endocytic uptake of a cell-penetrating peptide: the crucial role of cell culture protocols. Cytotechnology 2023; 75:473-490. [PMID: 37841959 PMCID: PMC10575844 DOI: 10.1007/s10616-023-00591-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 08/24/2023] [Indexed: 10/17/2023] Open
Abstract
Delivery tools, including cell-penetrating peptides (CPPs), are often inefficient due to a combination of poor endocytosis and endosomal escape. Aspects that impact the delivery of CPPs are typically characterized using tissue culture models. One problem of using cell culture is that cell culture protocols have the potential to contribute to endosomal uptake and endosomal release of CPPs. Hence, a systematic study to identify which aspects of cell culturing techniques impact the endocytic uptake of a typical CPP, the TMR-TAT peptide (peptide sequence derived from HIV1-TAT with the N-terminus labeled with tetramethylrhodamine), was conducted. Aspects of cell culturing protocols previously found to generally modulate endocytosis, such as cell density, washing steps, and cell aging, did not affect TMR-TAT endocytosis. In contrast, cell dissociation methods, media, temperature, serum starvation, and media composition all contributed to changes in uptake. To establish a range of endocytosis achievable by different cell culture protocols, TMR-TAT uptake was compared among protocols. These protocols led to changes in uptake of more than 13-fold, indicating that differences in cell culturing techniques have a cumulative effect on CPP uptake. Taken together this study highlights how different protocols can influence the amount of endocytic uptake of TMR-TAT. Additionally, parameters that can be exploited to improve CPP accumulation in endosomes were identified. The protocols identified herein have the potential to be paired with other delivery enhancing strategies to improve overall delivery efficiency of CPPs. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-023-00591-1.
Collapse
Affiliation(s)
- Joshua Diaz
- Department of Biochemistry and Biophysics, Texas A&M University, Room 430, 300 Olsen Blvd, College Station, TX 77843-2128 USA
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University, Room 430, 300 Olsen Blvd, College Station, TX 77843-2128 USA
- Department of Chemistry, Texas A&M University, College Station, TX 77843 USA
| |
Collapse
|
20
|
A Naveena H, Bhatia D. Hypoxia Modulates Cellular Endocytic Pathways and Organelles with Enhanced Cell Migration and 3D Cell Invasion. Chembiochem 2023; 24:e202300506. [PMID: 37677117 DOI: 10.1002/cbic.202300506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/09/2023]
Abstract
Hypoxia, a decrease in cellular or tissue level oxygen content, is characteristic of most tumors and has been shown to drive cancer progression by altering multiple subcellular processes. We hypothesized that the cancer cells in a hypoxic environment might have slower proliferation rates and increased invasion and migration rates with altered endocytosis compared to the cancer cells in the periphery of the tumor mass that experience normoxic conditions. We induced cellular hypoxia by exposing cells to cobalt chloride, a chemical hypoxic mimicking agent. This study measured the effect of hypoxia on cell proliferation, migration, and invasion. Uptake of fluorescently labeled transferrin, galectin3, and dextran that undergo endocytosis through major endocytic pathways (Clathrin-mediated pathway (CME), Clathrin-independent pathway (CIE), Fluid phase endocytosis (FPE)) were analyzed during hypoxia. Also, the organelle changes associated with hypoxia were studied with organelle trackers. We found that the proliferation rate decreased, and the migration and invasion rate increased in cancer cells in hypoxic conditions compared to normoxic cancer cells. A short hypoxic exposure increased galectin3 uptake in hypoxic cancer cells, but a prolonged hypoxic exposure decreased clathrin-independent endocytic uptake of galectin 3. Subcellular organelles, such as mitochondria, increased to withstand the hypoxic stress, while other organelles, such as Endoplasmic reticulum (ER), were significantly decreased. These data suggest that hypoxia modulates cellular endocytic pathways with reduced proliferation and enhanced cell migration and invasion.
Collapse
Affiliation(s)
- Hema A Naveena
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj Gandhinagar, 382355, Gujarat, India
| | - Dhiraj Bhatia
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj Gandhinagar, 382355, Gujarat, India
| |
Collapse
|
21
|
Espina JA, Cordeiro MH, Milivojevic M, Pajić-Lijaković I, Barriga EH. Response of cells and tissues to shear stress. J Cell Sci 2023; 136:jcs260985. [PMID: 37747423 PMCID: PMC10560560 DOI: 10.1242/jcs.260985] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023] Open
Abstract
Shear stress is essential for normal physiology and malignancy. Common physiological processes - such as blood flow, particle flow in the gut, or contact between migratory cell clusters and their substrate - produce shear stress that can have an impact on the behavior of different tissues. In addition, shear stress has roles in processes of biomedical interest, such as wound healing, cancer and fibrosis induced by soft implants. Thus, understanding how cells react and adapt to shear stress is important. In this Review, we discuss in vivo and in vitro data obtained from vascular and epithelial models; highlight the insights these have afforded regarding the general mechanisms through which cells sense, transduce and respond to shear stress at the cellular levels; and outline how the changes cells experience in response to shear stress impact tissue organization. Finally, we discuss the role of shear stress in collective cell migration, which is only starting to be appreciated. We review our current understanding of the effects of shear stress in the context of embryo development, cancer and fibrosis, and invite the scientific community to further investigate the role of shear stress in these scenarios.
Collapse
Affiliation(s)
- Jaime A. Espina
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| | - Marilia H. Cordeiro
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| | - Milan Milivojevic
- Faculty of Technology and Metallurgy, Belgrade University, 11120 Belgrade, Serbia
| | | | - Elias H. Barriga
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| |
Collapse
|
22
|
Sorrin AJ, Zhou K, May K, Liu C, McNaughton K, Rahman I, Liang BJ, Rizvi I, Roque DM, Huang HC. Transient fluid flow improves photoimmunoconjugate delivery and photoimmunotherapy efficacy. iScience 2023; 26:107221. [PMID: 37520715 PMCID: PMC10372742 DOI: 10.1016/j.isci.2023.107221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/01/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023] Open
Abstract
Circulating drugs in the peritoneal cavity is an effective strategy for advanced ovarian cancer treatment. Photoimmunotherapy, an emerging modality with potential for the treatment of ovarian cancer, involves near-infrared light activation of antibody-photosensitizer conjugates (photoimmunoconjugates) to generate cytotoxic reactive oxygen species. Here, a microfluidic cell culture model is used to study how fluid flow-induced shear stress affects photoimmunoconjugate delivery to ovarian cancer cells. Photoimmunoconjugates are composed of the antibody, cetuximab, conjugated to the photosensitizer, and benzoporphyrin derivative. Longitudinal tracking of photoimmunoconjugate treatment under flow conditions reveals enhancements in subcellular photosensitizer accumulation. Compared to static conditions, fluid flow-induced shear stress at 0.5 and 1 dyn/cm2 doubled the cellular delivery of photoimmunoconjugates. Fluid flow-mediated treatment with three different photosensitizer formulations (benzoporphyrin derivative, photoimmunoconjugates, and photoimmunoconjugate-coated liposomes) led to enhanced phototoxicity compared to static conditions. This study confirms the fundamental role of fluid flow-induced shear stress in the anti-cancer effects of photoimmunotherapy.
Collapse
Affiliation(s)
- Aaron J. Sorrin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Keri Zhou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Katherine May
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Cindy Liu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Kathryn McNaughton
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Idrisa Rahman
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Barry J. Liang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, North Carolina State University, Raleigh, NC 27599, USA
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dana M. Roque
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
23
|
Padovani-Claudio DA, Ramos CJ, Capozzi ME, Penn JS. Elucidating glial responses to products of diabetes-associated systemic dyshomeostasis. Prog Retin Eye Res 2023; 94:101151. [PMID: 37028118 PMCID: PMC10683564 DOI: 10.1016/j.preteyeres.2022.101151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 04/08/2023]
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness in working age adults. DR has non-proliferative stages, characterized in part by retinal neuroinflammation and ischemia, and proliferative stages, characterized by retinal angiogenesis. Several systemic factors, including poor glycemic control, hypertension, and hyperlipidemia, increase the risk of DR progression to vision-threatening stages. Identification of cellular or molecular targets in early DR events could allow more prompt interventions pre-empting DR progression to vision-threatening stages. Glia mediate homeostasis and repair. They contribute to immune surveillance and defense, cytokine and growth factor production and secretion, ion and neurotransmitter balance, neuroprotection, and, potentially, regeneration. Therefore, it is likely that glia orchestrate events throughout the development and progression of retinopathy. Understanding glial responses to products of diabetes-associated systemic dyshomeostasis may reveal novel insights into the pathophysiology of DR and guide the development of novel therapies for this potentially blinding condition. In this article, first, we review normal glial functions and their putative roles in the development of DR. We then describe glial transcriptome alterations in response to systemic circulating factors that are upregulated in patients with diabetes and diabetes-related comorbidities; namely glucose in hyperglycemia, angiotensin II in hypertension, and the free fatty acid palmitic acid in hyperlipidemia. Finally, we discuss potential benefits and challenges associated with studying glia as targets of DR therapeutic interventions. In vitro stimulation of glia with glucose, angiotensin II and palmitic acid suggests that: 1) astrocytes may be more responsive than other glia to these products of systemic dyshomeostasis; 2) the effects of hyperglycemia on glia are likely to be largely osmotic; 3) fatty acid accumulation may compound DR pathophysiology by promoting predominantly proinflammatory and proangiogenic transcriptional alterations of macro and microglia; and 4) cell-targeted therapies may offer safer and more effective avenues for DR treatment as they may circumvent the complication of pleiotropism in retinal cell responses. Although several molecules previously implicated in DR pathophysiology are validated in this review, some less explored molecules emerge as potential therapeutic targets. Whereas much is known regarding glial cell activation, future studies characterizing the role of glia in DR and how their activation is regulated and sustained (independently or as part of retinal cell networks) may help elucidate mechanisms of DR pathogenesis and identify novel drug targets for this blinding disease.
Collapse
Affiliation(s)
- Dolly Ann Padovani-Claudio
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, B3321A Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| | - Carla J Ramos
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, AA1324 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| | - Megan E Capozzi
- Duke Molecular Physiology Institute, Duke University School of Medicine, 300 North Duke Street, Durham, NC, 27701, USA.
| | - John S Penn
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, B3307 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| |
Collapse
|
24
|
Koller A, Brunner SM, Preishuber-Pflügl J, Runge C, Ladek AM, Reitsamer HA, Trost A. Cysteinyl leukotriene receptor 1 is a potent regulator of the endosomal-lysosomal system in the ARPE-19 retinal pigment epithelial cell line. Traffic 2023; 24:177-189. [PMID: 36704929 DOI: 10.1111/tra.12881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/28/2023]
Abstract
The endosomal-lysosomal system is central for cell homeostasis and comprises the functions and dynamics of particular organelles including endosomes, lysosomes and autophagosomes. In previous studies, we found that the cysteinyl leukotriene receptor 1 (CysLTR1) regulates autophagy in the retinal pigment epithelial cell line ARPE-19 under basal cellular conditions. However, the underlying mechanism by which CysLTR1 regulates autophagy is unknown. Thus, in the present study, the effects of CysLTR1 inhibition on the endosomal-lysosomal system are analyzed in detail to identify the role of CysLTR1 in cell homeostasis and autophagy regulation. CysLTR1 inhibition in ARPE-19 cells by Zafirlukast, a CysLTR1 antagonist, depleted the lysosomal pool. Furthermore, CysLTR1 antagonization reduced endocytic capacity and internalization of epidermal growth factor and decreased levels of the transferrin receptor, CD71. Serum starvation abolished the effect of Zafirlukast on the autophagic flux, which identifies the endocytic regulation of serum components by CysLTR1 as an important autophagy-modulating mechanism. The role of CysLTR1 in inflammation and cell stress has been exceedingly studied, but its involvement in the endosomal-lysosomal pathway is largely unknown. This current study provides new insights into basal activity of CysLTR1 on cellular endocytosis and the subsequent impact on downstream processes like autophagy.
Collapse
Affiliation(s)
- Andreas Koller
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Susanne Maria Brunner
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Julia Preishuber-Pflügl
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Christian Runge
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Anja-Maria Ladek
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Herbert Anton Reitsamer
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Andrea Trost
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
25
|
Rahmani S, Ahmed H, Ibazebo O, Fussner-Dupas E, Wakarchuk WW, Antonescu CN. O-GlcNAc transferase modulates the cellular endocytosis machinery by controlling the formation of clathrin-coated pits. J Biol Chem 2023; 299:102963. [PMID: 36731797 PMCID: PMC9999237 DOI: 10.1016/j.jbc.2023.102963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 02/01/2023] Open
Abstract
Clathrin-mediated endocytosis (CME) controls the internalization and function of a wide range of cell surface proteins. CME occurs by the assembly of clathrin and many other proteins on the inner leaflet of the plasma membrane into clathrin-coated pits (CCPs). These structures recruit specific cargo destined for internalization, generate membrane curvature, and in many cases undergo scission from the plasma membrane to yield intracellular vesicles. The diversity of functions of cell surface proteins controlled via internalization by CME may suggest that regulation of CCP formation could be effective to allow cellular adaptation under different contexts. Of interest is how cues derived from cellular metabolism may regulate CME, given the reciprocal role of CME in controlling cellular metabolism. The modification of proteins with O-linked β-GlcNAc (O-GlcNAc) is sensitive to nutrient availability and may allow cellular adaptation to different metabolic conditions. Here, we examined how the modification of proteins with O-GlcNAc may control CCP formation and thus CME. We used perturbation of key enzymes responsible for protein O-GlcNAc modification, as well as specific mutants of the endocytic regulator AAK1 predicted to be impaired for O-GlcNAc modification. We identify that CCP initiation and the assembly of clathrin and other proteins within CCPs are controlled by O-GlcNAc protein modification. This reveals a new dimension of regulation of CME and highlights the important reciprocal regulation of cellular metabolism and endocytosis.
Collapse
Affiliation(s)
- Sadia Rahmani
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Hafsa Ahmed
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Osemudiamen Ibazebo
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Eden Fussner-Dupas
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Warren W Wakarchuk
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.
| | - Costin N Antonescu
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada.
| |
Collapse
|
26
|
Gilthead Seabream Liver Integrative Proteomics and Metabolomics Analysis Reveals Regulation by Different Prosurvival Pathways in the Metabolic Adaptation to Stress. Int J Mol Sci 2022; 23:ijms232315395. [PMID: 36499720 PMCID: PMC9741202 DOI: 10.3390/ijms232315395] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
The study of the molecular mechanisms of stress appraisal on farmed fish is paramount to ensuring a sustainable aquaculture. Stress exposure can either culminate in the organism's adaptation or aggravate into a metabolic shutdown, characterized by irreversible cellular damage and deleterious effects on fish performance, welfare, and survival. Multiomics can improve our understanding of the complex stressed phenotype in fish and the molecular mediators that regulate the underlying processes of the molecular stress response. We profiled the stress proteome and metabolome of Sparus aurata responding to different challenges common to aquaculture production, characterizing the disturbed pathways in the fish liver, i.e., the central organ in mounting the stress response. Label-free shotgun proteomics and untargeted metabolomics analyses identified 1738 proteins and 120 metabolites, separately. Mass spectrometry data have been made fully accessible via ProteomeXchange, with the identifier PXD036392, and via MetaboLights, with the identifier MTBLS5940. Integrative multivariate statistical analysis, performed with data integration analysis for biomarker discovery using latent components (DIABLO), depicted the 10 most-relevant features. Functional analysis of these selected features revealed an intricate network of regulatory components, modulating different signaling pathways related to cellular stress, e.g., the mTORC1 pathway, the unfolded protein response, endocytosis, and autophagy to different extents according to the stress nature. These results shed light on the dynamics and extent of this species' metabolic reprogramming under chronic stress, supporting future studies on stress markers' discovery and fish welfare research.
Collapse
|
27
|
Dong Y, Jia R, Hou Y, Diao W, Li B, Zhu J. Effects of stocking density on the growth performance, mitophagy, endocytosis and metabolism of Cherax quadricarinatus in integrated rice-crayfish farming systems. Front Physiol 2022; 13:1040712. [PMID: 36518112 PMCID: PMC9742548 DOI: 10.3389/fphys.2022.1040712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/16/2022] [Indexed: 07/30/2023] Open
Abstract
Red claw crayfish (Cherax quadricarinatus) is an economic freshwater shrimp with great commercial potential. However, the suitable stocking density of C. quadricarinatus is still unclear in integrated rice-crayfish farming system. Thus, this study aimed to investigate the effects of stocking density on growth performance, mitophagy, endocytosis and metabolism of C. quadricarinatus. The C. quadricarinatus was reared at low density (LD, 35.73 g/m2), middle density (MD, 71.46 g/m2) and high density (HD, 107.19 g/m2) in an integrated rice-crayfish farming system. After 90 days of farming, the growth performance of C. quadricarinatus significantly decreased in the MD and HD groups relative to that in the LD group. The HD treatment caused oxidative stress and lipid peroxidation at the end of the experiment in hepatopancreas. Transcriptome analysis showed that there were 1,531 DEGs (differently expressed genes) between the LD group and HD group, including 1,028 upregulated genes and 503 downregulated genes. KEGG (Kyoto Encyclopedia of Genes and Genomes) enrichment analysis indicated that the DEGs were significantly enriched in endocytosis and mitophagy pathways. Meanwhile, four lipid metabolism pathways, including biosynthesis of unsaturated fatty acids, fatty acid biosynthesis, glycerolipid metabolism and glycerophospholipid metabolism, exhibited an upregulated tendency in the HD group. In conclusion, our data showed that when the stocking density reached up to 207.15 g/m2 in HD group, the growth performance of C. quadricarinatus was significantly inhibited in this system. Meanwhile, the data indicated that C. quadricarinatus may respond to the stressful condition via activating antioxidant defense system, endocytosis, mitophagy and metabolism-related pathways in hepatopancreas.
Collapse
Affiliation(s)
- Yin Dong
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Rui Jia
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Yiran Hou
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Weixu Diao
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Bing Li
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Jian Zhu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| |
Collapse
|
28
|
Nano Modification of Antrodia Cinnamomea Exhibits Anti-Inflammatory Action and Improves the Migratory Potential of Myogenic Progenitors. Cells 2022; 11:cells11162512. [PMID: 36010589 PMCID: PMC9406806 DOI: 10.3390/cells11162512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/21/2022] Open
Abstract
The skeletal muscle progenitors’ proliferation and migration are crucial stages of myogenesis. Identifying drug candidates that contribute to myogenesis can have a positive impact on atrophying muscle. The purpose of the study is to synthesize the Antrodia cinnamomea (AC)-β-cyclodextrin (BCD) inclusion complex (IC) and understand its in vitro pro-regenerative influence in murine skeletal C2C12 myoblasts. The IC was subjected to various nano-characterization studies. Fluorescent IC was synthesized to understand the cellular uptake of IC. Furthermore, 25 µg/mL, 12.5 µg/mL, and 6.25 µg/mL of IC were tested on murine C2C12 skeletal muscle cells for their anti-inflammatory, pro-migratory, and pro-proliferative action. The cellular internalization of IC occurred rapidly via pinocytosis. IC (252.6 ± 3.2 nm size and −37.24 ± 1.55 surface charge) exhibited anti-inflammatory action by suppressing the secretion of interleukin-6 and enhanced cell proliferation with promising cytocompatibility. A 12.5 μg/mL dose of IC promoted cell migration in 24 h, but the same dose of AC significantly reduced cell migration, suggesting modification by BCD. Molecular studies revealed that IC promoted C2C12 myoblasts migration by upregulating long non-coding RNA (lncRNA) NEAT-1, SYISL, and activating the pPKC/β-catenin pathway. Our study is the first report on the pro-proliferative and pro-migratory effects of BCD-modified extracts of AC.
Collapse
|
29
|
Barbeau S, Joushomme A, Chappe Y, Cardouat G, Baudrimont I, Freund-Michel V, Guibert C, Marthan R, Berger P, Vacher P, Percherancier Y, Quignard JF, Ducret T. Cell Confluence Modulates TRPV4 Channel Activity in Response to Hypoxia. Biomolecules 2022; 12:954. [PMID: 35883510 PMCID: PMC9313184 DOI: 10.3390/biom12070954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a polymodal Ca2+-permeable channel involved in various hypoxia-sensitive pathophysiological phenomena. Different tools are available to study channel activity, requiring cells to be cultured at specific optimal densities. In the present study, we examined if cell density may influence the effect of hypoxia on TRPV4 activity. Transiently TRPV4-transfected HEK293T cells were seeded at low or high densities corresponding to non-confluent or confluent cells, respectively, on the day of experiments, and cultured under in vitro normoxia or hypoxia. TRPV4-mediated cytosolic Ca2+ responses, single-channel currents, and Ca2+ influx through the channel were measured using Ca2+ imaging/microspectrofluorimetric assay, patch-clamp, and Bioluminescence Resonance Energy Transfer (BRET), respectively. TRPV4 plasma membrane translocation was studied using confocal microscopy, biotinylation of cell surface proteins, and BRET. Our results show that hypoxia exposure has a differential effect on TRPV4 activation depending on cell confluence. At low confluence levels, TRPV4 response is increased in hypoxia, whereas at high confluence levels, TRPV4 response is strongly inhibited, due to channel internalization. Thus, cell density appears to be a crucial parameter for TRPV4 channel activity.
Collapse
Affiliation(s)
- Solène Barbeau
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Alexandre Joushomme
- Laboratoire de l’Intégration du Matériau au Système, UMR5518, Univ. Bordeaux, F-33400 Talence, France; (A.J.); (Y.C.); (Y.P.)
- CNRS (Centre National de la Recherche Scientifique), Laboratoire de L’integration du Matériau au Système, UMR5518, F-33400 Talence, France
| | - Yann Chappe
- Laboratoire de l’Intégration du Matériau au Système, UMR5518, Univ. Bordeaux, F-33400 Talence, France; (A.J.); (Y.C.); (Y.P.)
- CNRS (Centre National de la Recherche Scientifique), Laboratoire de L’integration du Matériau au Système, UMR5518, F-33400 Talence, France
| | - Guillaume Cardouat
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Isabelle Baudrimont
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Véronique Freund-Michel
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Christelle Guibert
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Roger Marthan
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
- CHU (Centre Hospitalier Universitaire) Bordeaux, Service d’Exploration Fonctionnelle Respiratoire, F-33600 Pessac, France
| | - Patrick Berger
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
- CHU (Centre Hospitalier Universitaire) Bordeaux, Service d’Exploration Fonctionnelle Respiratoire, F-33600 Pessac, France
| | - Pierre Vacher
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Yann Percherancier
- Laboratoire de l’Intégration du Matériau au Système, UMR5518, Univ. Bordeaux, F-33400 Talence, France; (A.J.); (Y.C.); (Y.P.)
- CNRS (Centre National de la Recherche Scientifique), Laboratoire de L’integration du Matériau au Système, UMR5518, F-33400 Talence, France
| | - Jean-François Quignard
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Thomas Ducret
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| |
Collapse
|
30
|
Nishimura A, Nakagami K, Kan K, Morita F, Takagi H. Arginine inhibits Saccharomyces cerevisiae biofilm formation by inducing endocytosis of the arginine transporter Can1. Biosci Biotechnol Biochem 2022; 86:1300-1307. [PMID: 35749478 DOI: 10.1093/bbb/zbac094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022]
Abstract
Biofilms are formed by the aggregation of microorganisms into multicellular structures that adhere to surfaces. Biofilm formation by yeast is a critical issue in clinical and industrial fields because of the strong adhesion of yeast biofilm to abiotic surfaces and tissues. Here, we clarified the arginine-mediated inhibition of biofilm formation by yeast. First, we showed that arginine inhibits biofilm formation in fungi such as Saccharomyces cerevisiae, Candida glabrata, and Cladosporium cladosporioides, but not in bacteria. In regard to the underlying mechanism, biochemical analysis indicated that arginine inhibits biofilm formation by suppressing Flo11-dependent flocculation. Intriguingly, a strain with deletion of the arginine transporter-encoding CAN1 was insensitive to arginine-mediated inhibition of biofilm formation. Finally, Can1 endocytosis appeared to be required for the inhibitory mechanism of biofilm formation by arginine. The present results could help to elucidate the molecular mechanism of yeast biofilm formation and its control.
Collapse
Affiliation(s)
- Akira Nishimura
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, Japan
| | - Kazuki Nakagami
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, Japan
| | - Kyoyuki Kan
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, Japan
| | - Fumika Morita
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, Japan
| | - Hiroshi Takagi
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, Japan
| |
Collapse
|
31
|
Dahhan DA, Bednarek SY. Advances in structural, spatial, and temporal mechanics of plant endocytosis. FEBS Lett 2022; 596:2269-2287. [PMID: 35674447 DOI: 10.1002/1873-3468.14420] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 11/06/2022]
Abstract
Endocytic trafficking underlies processes essential for plant growth and development, including the perception of and response to abiotic and extracellular stimuli, post-Golgi and exocytic trafficking, and cytokinesis. Protein adaptors and regulatory factors of clathrin-mediated endocytosis that contribute to the formation of endocytic clathrin-coated vesicles are evolutionarily conserved. Yet, work of the last ten years has identified differences between the endocytic mechanisms of plants and Opisthokonts involving the endocytic adaptor TPLATE complex, the requirement of actin during CME, and the function of clathrin-independent endocytosis in the uptake of plant-specific plasma membrane proteins. Here, we review clathrin-mediated and -independent pathways in plants and describe recent advances enabled by new proteomic and imaging methods, and conditional perturbation of endocytosis. In addition, we summarize the formation and trafficking of clathrin-coated vesicles based on temporal and structural data garnered from high-resolution quantitative imaging studies. Finally, new information about the cross-talk between endocytosis and other endomembrane trafficking pathways and organelles will also be discussed.
Collapse
Affiliation(s)
- Dana A Dahhan
- Department of Biochemistry, University of Wisconsin-Madison, WI, USA
| | | |
Collapse
|
32
|
Xiong P, Cheng XY, Sun XY, Chen XW, Ouyang JM. Interaction between nanometer calcium oxalate and renal epithelial cells repaired with carboxymethylated polysaccharides. BIOMATERIALS ADVANCES 2022; 137:212854. [PMID: 35929244 DOI: 10.1016/j.bioadv.2022.212854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/06/2022] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE Injury of renal tubular epithelial cells (HK-2) is an important cause of kidney stone formation. In this article, the repairing effect of polysaccharide (PCP0) extracted from the traditional Chinese medicine Poria cocos and its carboxymethylated derivatives on damaged HK-2 cells was studied, and the differences in adhesion and endocytosis of the cells to nanometer calcium oxalate monohydrate (COM) before and after repair were explored. METHODS Sodium oxalate (2.8 mmol/L) was used to damage HK-2 cells to establish a damage model, and then Poria cocos polysaccharides (PCPs) with different carboxyl (COOH) contents were used to repair the damaged cells. The changes in the biochemical indicators of the cells before and after the repair and the changes in the ability to adhere to and internalize nano-COM were detected. RESULTS The natural PCPs (PCP0, COOH content = 2.56%) were carboxymethylated, and three carboxylated modified Poria cocos with 7.48% (PCP1), 12.07% (PCP2), and 17.18% (PCP3) COOH contents were obtained. PCPs could repair the damaged HK-2 cells, and the cell viability was enhanced after repair. The cell morphology was gradually repaired, the proliferation and healing rate were increased. The ROS production was reduced, and the polarity of the mitochondrial membrane potential was restored. The level of intracellular Ca2+ ions decreased, and the autophagy response was weakened. CONCLUSION The cells repaired by PCPs inhibited the adhesion to nano-COM and simultaneously promoted the endocytosis of nano-COM. The endocytic crystals mainly accumulated in the lysosome. Inhibiting adhesion and increasing endocytosis could reduce the nucleation, growth, and aggregation of cell surface crystals, thereby inhibiting the formation of kidney stones. With the increase of COOH content in PCPs, its ability to repair damaged cells, inhibit crystal adhesion, and promote crystal endocytosis all increased, that is, PCP3 with the highest COOH content showed the best ability to inhibit stone formation.
Collapse
Affiliation(s)
- Peng Xiong
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| | - Xiao-Yan Cheng
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| | - Xin-Yuan Sun
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Xue-Wu Chen
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| | - Jian-Ming Ouyang
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
33
|
Petrov AM, Zakirjanova GF, Kovyazina IV, Tsentsevitsky AN, Bukharaeva EA. Adrenergic receptors control frequency-dependent switching of the exocytosis mode between "full-collapse" and "kiss-and-run" in murine motor nerve terminal. Life Sci 2022; 296:120433. [PMID: 35219696 DOI: 10.1016/j.lfs.2022.120433] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 12/22/2022]
Abstract
AIMS Neurotransmitter release from the synaptic vesicles can occur through two modes of exocytosis: "full-collapse" or "kiss-and-run". Here we investigated how increasing the nerve activity and pharmacological stimulation of adrenoceptors can influence the mode of exocytosis in the motor nerve terminal. METHODS Recording of endplate potentials with intracellular microelectrodes was used to estimate acetylcholine release. A fluorescent dye FM1-43 and its quenching with sulforhodamine 101 were utilized to visualize synaptic vesicle recycling. KEY FINDINGS An increase in the frequency of stimulation led to a decrease in the rate of FM1-43 unloading despite the higher number of quanta released. High frequency activity promoted neurotransmitter release via the kiss-and-run mechanism. This was confirmed by experiments utilizing (I) FM1-43 dye quencher, that is able to pass into the synaptic vesicle via fusion pore, and (II) loading of FM1-43 by compensatory endocytosis. Noradrenaline and specific α2-adrenoreceptors agonist, dexmedetomidine, controlled the mode of synaptic vesicle recycling at high frequency activity. Their applications favored neurotransmitter release via full-collapse exocytosis rather than the kiss-and-run pathway. SIGNIFICANCE At the diaphragm neuromuscular junctions, neuronal commands are translated into contractions necessary for respiration. During stress, an increase in discharge rate of the phrenic nerve shifts the exocytosis from the full-collapse to the kiss-and-run mode. The stress-related molecule, noradrenaline, restricts neurotransmitter release in response to a high frequency activity, and prevents the shift in the mode of exocytosis through α2-adrenoceptor activation. This may be a component of the mechanism that limits overstimulation of the respiratory system during stress.
Collapse
Affiliation(s)
- Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, Russia; Kazan State Medical University, Kazan, Russia.
| | - Guzalia F Zakirjanova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, Russia
| | - Irina V Kovyazina
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, Russia; Kazan State Medical University, Kazan, Russia
| | - Andrei N Tsentsevitsky
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, Russia
| | - Ellya A Bukharaeva
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, Russia
| |
Collapse
|
34
|
Shi W, Hu R, Wang P, Zhao R, Shen H, Li H, Wang L, Qiao Y, Jiang G, Cheng J, Wan X. Transcriptome analysis of acute high temperature-responsive genes and pathways in Palaemon gravieri. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2022; 41:100958. [PMID: 34999569 DOI: 10.1016/j.cbd.2021.100958] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 10/19/2022]
Abstract
Temperature is an important variable factor in aquaculture which affects the health, survival, behavior, growth, and development of aquatic animals. Palaemon gravieri is one of the main economic shrimps in marine capture fisheries of the East China Sea and the South China Yellow Sea; however, it cannot tolerate high temperatures, thereby, resulting in unsuccessful large-scale farming. Thus far, there are few studies on the effects of acute high temperature on P. graviera. Therefore, it is especially important to study the effects of temperature fluctuations, especially acute high temperature, on P. gravieri. In this study, P. gravieri was treated with acute high-temperature stress, which gradually rose from 15 °C to 30 °C in 3 h, then remained at 30 °C for 12 h. The hepatopancreas of shrimps from five time points was collected once at 15 °C and thereafter, every 3 h after 30 °C. The samples of G0, G1, and G4 were selected for transcriptome analysis. A total of 18,308 unigenes were annotated, of which 7744 were differentially expressed. Most differentially expressed genes (DEGs) come from several physiological and biochemical processes, such as metabolism (GRHPR, ALDH5A1, GDH), immunity (HSP70, Rab5B, Rab10, CASP7), and stress-related process (UGT, GST, HSP60, HSP90). The results indicated that acute high temperature significantly reduced the metabolic capacity of shrimp but enhanced the immune capacity, which seemed to be an emergency metabolic compensation technique to resist stress. This study contributes to ongoing research on the physiological mechanism of P. gravieri response to acute high temperature.
Collapse
Affiliation(s)
- Wenjun Shi
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China; Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China
| | - Runhao Hu
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Pan Wang
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Ran Zhao
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Hui Shen
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China
| | - Hui Li
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China
| | - Libao Wang
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China
| | - Yi Qiao
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China
| | - Ge Jiang
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China
| | - Jie Cheng
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China
| | - Xihe Wan
- Institute of Oceanology & Marine Fisheries, Jiangsu, Nantong, China.
| |
Collapse
|
35
|
Exposure of Caenorhabditis elegans to Dietary Nε-Carboxymethyllysine Emphasizes Endocytosis as a New Route for Intestinal Absorption of Advanced Glycation End Products. Nutrients 2021; 13:nu13124398. [PMID: 34959950 PMCID: PMC8705817 DOI: 10.3390/nu13124398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/11/2022] Open
Abstract
The impact of dietary advanced glycation end products (dAGEs) on human health has been discussed in many studies but, to date, no consensual pathophysiological process has been demonstrated. The intestinal absorption pathways which have so far been described for dAGEs, the passive diffusion of free AGE adducts and transport of glycated di-tripeptides by the peptide transporter 1 (PEPT-1), are not compatible with certain pathophysiological processes described. To get new insight into the intestinal absorption pathways and the pathophysiological mechanisms of dAGEs, we initiated an in vivo study with a so-called simple animal model with a complete digestive tract, Caenorhabditis elegans. Dietary bacteria were chemically modified with glyoxylic acid to mainly produce Nε-carboxymethyllysine (CML) and used to feed the worms. We performed different immunotechniques using an anti-CML antibody for the relative quantification of ingested CML and localization of this AGE in the worms’ intestine. The relative expression of genes encoding different biological processes such as response to stresses and intestinal digestion were determined. The physiological development of the worms was verified. All the results were compared with those obtained with the control bacteria. The results revealed a new route for the intestinal absorption of dietary CML (dCML), endocytosis, which could be mediated by scavenger receptors. The exposure of worms to dCML induced a reproductive defect and a transcriptional response reflecting oxidative, carbonyl and protein folding stresses. These data, in particular the demonstration of endocytosis of dCML by enterocytes, open up new perspectives to better characterize the pathophysiological mechanisms of dAGEs.
Collapse
|
36
|
Both Clathrin-Mediated and Membrane Microdomain-Associated Endocytosis Contribute to the Cellular Adaptation to Hyperosmotic Stress in Arabidopsis. Int J Mol Sci 2021; 22:ijms222212534. [PMID: 34830417 PMCID: PMC8621756 DOI: 10.3390/ijms222212534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 11/23/2022] Open
Abstract
As sessile organisms, plants must directly deal with an often complex and adverse environment in which hyperosmotic stress is one of the most serious abiotic factors, challenging cellular physiology and integrity. The plasma membrane (PM) is the hydrophobic barrier between the inside and outside environments of cells and is considered a central compartment in cellular adaptation to diverse stress conditions through dynamic PM remodeling. Endocytosis is a powerful method for rapid remodeling of the PM. In animal cells, different endocytic pathways are activated in response to osmotic stress, while only a few reports are related to the endocytosis response pathway and involve a mechanism in plant cells upon hyperosmotic stress. In this study, using different endocytosis inhibitors, the microdomain-specific dye di-4-ANEPPDHQ, variable-angle total internal reflection fluorescence microscopy (VA-TIRFM), and confocal microscopy, we discovered that internalized Clathrin Light Chain-Green Fluorescent Protein (CLC-GFP) increased under hyperosmotic conditions, accompanied by decreased fluorescence intensity of CLC-GFP at the PM. CLC-GFP tended to have higher diffusion coefficients and a fraction of CLC-GFP molecules underwent slower diffusion upon hyperosmotic stress. Meanwhile, an increased motion range of CLC-GFP was found under hyperosmotic treatment compared with the control. In addition, the order of the PM decreased, but the order of the endosome increased when cells were in hyperosmotic conditions. Hence, our results demonstrated that clathrin-mediated endocytosis and membrane microdomain-associated endocytosis both participate in the adaptation to hyperosmotic stress. These findings will help to further understand the role and the regulatory mechanism involved in plant endocytosis in helping plants adapt to osmotic stress.
Collapse
|
37
|
Influence of formic acid treatment on the proteome of the ectoparasite Varroa destructor. PLoS One 2021; 16:e0258845. [PMID: 34699527 PMCID: PMC8547630 DOI: 10.1371/journal.pone.0258845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 10/06/2021] [Indexed: 11/21/2022] Open
Abstract
The ectoparasite Varroa destructor Anderson and Trueman is the most important parasites of the western honey bee, Apis mellifera L. The most widely currently used treatment uses formic acid (FA), but the understanding of its effects on V. destructor is limited. In order to understand the mechanism of action of FA, its effect on Varroa mites was investigated using proteomic analysis by liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). V. destructor was collected from honey bee colonies with natural mite infestation before and 24 h after the initiation of FA treatment and subjected to proteome analysis. A total of 2637 proteins were identified. Quantitative analysis of differentially expressed candidate proteins (fold change ≥ 1.5; p ≤ 0.05) revealed 205 differentially expressed proteins: 91 were induced and 114 repressed in the FA-treated group compared to the untreated control group. Impaired protein synthesis accompanied by increased protein and amino acid degradation suggest an imbalance in proteostasis. Signs of oxidative stress included significant dysregulation of candidate proteins of mitochondrial cellular respiration, increased endocytosis, and induction of heat shock proteins. Furthermore, an increased concentration of several candidate proteins associated with detoxification was observed. These results suggest dysregulated cellular respiration triggered by FA treatment as well as an increase in cellular defense mechanisms, including induced heat shock proteins and detoxification enzymes.
Collapse
|
38
|
Bonilla DA, Moreno Y, Rawson ES, Forero DA, Stout JR, Kerksick CM, Roberts MD, Kreider RB. A Convergent Functional Genomics Analysis to Identify Biological Regulators Mediating Effects of Creatine Supplementation. Nutrients 2021; 13:2521. [PMID: 34444681 PMCID: PMC8397972 DOI: 10.3390/nu13082521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Creatine (Cr) and phosphocreatine (PCr) are physiologically essential molecules for life, given they serve as rapid and localized support of energy- and mechanical-dependent processes. This evolutionary advantage is based on the action of creatine kinase (CK) isozymes that connect places of ATP synthesis with sites of ATP consumption (the CK/PCr system). Supplementation with creatine monohydrate (CrM) can enhance this system, resulting in well-known ergogenic effects and potential health or therapeutic benefits. In spite of our vast knowledge about these molecules, no integrative analysis of molecular mechanisms under a systems biology approach has been performed to date; thus, we aimed to perform for the first time a convergent functional genomics analysis to identify biological regulators mediating the effects of Cr supplementation in health and disease. A total of 35 differentially expressed genes were analyzed. We identified top-ranked pathways and biological processes mediating the effects of Cr supplementation. The impact of CrM on miRNAs merits more research. We also cautiously suggest two dose-response functional pathways (kinase- and ubiquitin-driven) for the regulation of the Cr uptake. Our functional enrichment analysis, the knowledge-based pathway reconstruction, and the identification of hub nodes provide meaningful information for future studies. This work contributes to a better understanding of the well-reported benefits of Cr in sports and its potential in health and disease conditions, although further clinical research is needed to validate the proposed mechanisms.
Collapse
Affiliation(s)
- Diego A. Bonilla
- Research Division, Dynamical Business & Science Society—DBSS International SAS, Bogotá 110861, Colombia;
- Research Group in Biochemistry and Molecular Biology, Universidad Distrital Francisco José de Caldas, Bogotá 110311, Colombia
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, Montería 230002, Colombia
- kDNA Genomics, Joxe Mari Korta Research Center, University of the Basque Country UPV/EHU, 20018 Donostia-San Sebastián, Spain
| | - Yurany Moreno
- Research Division, Dynamical Business & Science Society—DBSS International SAS, Bogotá 110861, Colombia;
- Research Group in Biochemistry and Molecular Biology, Universidad Distrital Francisco José de Caldas, Bogotá 110311, Colombia
| | - Eric S. Rawson
- Department of Health, Nutrition and Exercise Science, Messiah University, Mechanicsburg, PA 17055, USA;
| | - Diego A. Forero
- Professional Program in Sport Training, School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá 111221, Colombia;
| | - Jeffrey R. Stout
- Physiology of Work and Exercise Response (POWER) Laboratory, Institute of Exercise Physiology and Rehabilitation Science, University of Central Florida, Orlando, FL 32816, USA;
| | - Chad M. Kerksick
- Exercise and Performance Nutrition Laboratory, School of Health Sciences, Lindenwood University, Saint Charles, MO 63301, USA;
| | - Michael D. Roberts
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA;
- Edward via College of Osteopathic Medicine, Auburn, AL 36849, USA
| | - Richard B. Kreider
- Exercise & Sport Nutrition Laboratory, Human Clinical Research Facility, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
39
|
Braz CU, Rowan TN, Schnabel RD, Decker JE. Genome-wide association analyses identify genotype-by-environment interactions of growth traits in Simmental cattle. Sci Rep 2021; 11:13335. [PMID: 34172761 PMCID: PMC8233360 DOI: 10.1038/s41598-021-92455-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
Understanding genotype-by-environment interactions (G × E) is crucial to understand environmental adaptation in mammals and improve the sustainability of agricultural production. Here, we present an extensive study investigating the interaction of genome-wide SNP markers with a vast assortment of environmental variables and searching for SNPs controlling phenotypic variance (vQTL) using a large beef cattle dataset. We showed that G × E contribute 10.1%, 3.8%, and 2.8% of the phenotypic variance of birth weight, weaning weight, and yearling weight, respectively. G × E genome-wide association analysis (GWAA) detected a large number of G × E loci affecting growth traits, which the traditional GWAA did not detect, showing that functional loci may have non-additive genetic effects regardless of differences in genotypic means. Further, variance-heterogeneity GWAA detected loci enriched with G × E effects without requiring prior knowledge of the interacting environmental factors. Functional annotation and pathway analysis of G × E genes revealed biological mechanisms by which cattle respond to changes in their environment, such as neurotransmitter activity, hypoxia-induced processes, keratinization, hormone, thermogenic and immune pathways. We unraveled the relevance and complexity of the genetic basis of G × E underlying growth traits, providing new insights into how different environmental conditions interact with specific genes influencing adaptation and productivity in beef cattle and potentially across mammals.
Collapse
Affiliation(s)
- Camila U Braz
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Troy N Rowan
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA
- Genetics Area Program, University of Missouri, Columbia, MO, 65211, USA
| | - Robert D Schnabel
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA
- Genetics Area Program, University of Missouri, Columbia, MO, 65211, USA
- Informatics Institute, University of Missouri, Columbia, MO, 65211, USA
| | - Jared E Decker
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA.
- Genetics Area Program, University of Missouri, Columbia, MO, 65211, USA.
- Informatics Institute, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
40
|
Svitkova B, Zavisova V, Nemethova V, Koneracka M, Kretova M, Razga F, Ursinyova M, Gabelova A. Differences in surface chemistry of iron oxide nanoparticles result in different routes of internalization. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:270-281. [PMID: 33842184 PMCID: PMC8008094 DOI: 10.3762/bjnano.12.22] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/11/2021] [Indexed: 06/12/2023]
Abstract
The efficient entry of nanotechnology-based pharmaceuticals into target cells is highly desired to reach high therapeutic efficiency while minimizing the side effects. Despite intensive research, the impact of the surface coating on the mechanism of nanoparticle uptake is not sufficiently understood yet. Herein, we present a mechanistic study of cellular internalization pathways of two magnetic iron oxide nanoparticles (MNPs) differing in surface chemistry into A549 cells. The MNP uptake was investigated in the presence of different inhibitors of endocytosis and monitored by spectroscopic and imaging techniques. The results revealed that the route of MNP entry into cells strongly depends on the surface chemistry of the MNPs. While serum bovine albumin-coated MNPs entered the cells via clathrin-mediated endocytosis (CME), caveolin-mediated endocytosis (CavME) or lipid rafts were preferentially involved in the internalization of polyethylene glycol-coated MNPs. Our data indicate that surface engineering can contribute to an enhanced delivery efficiency of nanoparticles.
Collapse
Affiliation(s)
- Barbora Svitkova
- Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Vlasta Zavisova
- Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 040 01 Kosice, Slovakia
| | - Veronika Nemethova
- Faculty of Medicine, Comenius University, Spitalska 24, 813 72 Bratislava, Slovakia
- Selecta Biotech SE, Istrijska 20, 841 07 Bratislava, Slovakia
| | - Martina Koneracka
- Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 040 01 Kosice, Slovakia
| | - Miroslava Kretova
- Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Filip Razga
- Faculty of Medicine, Comenius University, Spitalska 24, 813 72 Bratislava, Slovakia
- Selecta Biotech SE, Istrijska 20, 841 07 Bratislava, Slovakia
| | - Monika Ursinyova
- Slovak Medical University, Limbova 12, 833 03 Bratislava, Slovakia
| | - Alena Gabelova
- Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| |
Collapse
|
41
|
Kubohara Y, Homma Y, Shibata H, Oshima Y, Kikuchi H. Dictyostelium Differentiation-Inducing Factor-1 Promotes Glucose Uptake, at Least in Part, via an AMPK-Dependent Pathway in Mouse 3T3-L1 Cells. Int J Mol Sci 2021; 22:2293. [PMID: 33669058 PMCID: PMC7956221 DOI: 10.3390/ijms22052293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 11/16/2022] Open
Abstract
Differentiation-inducing factor-1 (DIF-1) is a chlorinated alkylphenone (a polyketide) found in the cellular slime mold Dictyostelium discoideum. DIF-1 and its derivative, DIF-1(3M) promote glucose consumption in vitro in mammalian cells and in vivo in diabetic rats; they are expected to be the leading antiobesity and antidiabetes compounds. In this study, we investigated the mechanisms underlying the actions of DIF-1 and DIF-1(3M). In isolated mouse liver mitochondria, these compounds at 2-20 μM promoted oxygen consumption in a dose-dependent manner, suggesting that they act as mitochondrial uncouplers, whereas CP-DIF-1 (another derivative of DIF-1) at 10-20 μM had no effect. In confluent mouse 3T3-L1 fibroblasts, DIF-1 and DIF-1(3M) but not CP-DIF-1 induced phosphorylation (and therefore activation) of AMP kinase (AMPK) and promoted glucose consumption and metabolism. The DIF-induced glucose consumption was reduced by compound C (an AMPK inhibitor) or AMPK knock down. These data suggest that DIF-1 and DIF-1(3M) promote glucose uptake, at least in part, via an AMPK-dependent pathway in 3T3-L1 cells, whereas cellular metabolome analysis revealed that DIF-1 and DIF-1(3M) may act differently at least in part.
Collapse
Affiliation(s)
- Yuzuru Kubohara
- Laboratory of Health and Life Science, Graduate School of Health and Sports Science, Juntendo University, Inzai, Chiba 270-1695, Japan
| | - Yoshimi Homma
- Department of Biomolecular Science, Institute of Biomedical Sciences, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan;
| | - Hiroshi Shibata
- Laboratory of Epigenetics and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan;
| | - Yoshiteru Oshima
- Laboratory of Natural Product Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aza-Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan; (Y.O.); (H.K.)
| | - Haruhisa Kikuchi
- Laboratory of Natural Product Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aza-Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan; (Y.O.); (H.K.)
| |
Collapse
|
42
|
Gammella E, Lomoriello IS, Conte A, Freddi S, Alberghini A, Poli M, Sigismund S, Cairo G, Recalcati S. Unconventional endocytosis and trafficking of transferrin receptor induced by iron. Mol Biol Cell 2020; 32:98-108. [PMID: 33236955 PMCID: PMC8120689 DOI: 10.1091/mbc.e20-02-0129] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The posttranslational regulation of transferrin receptor (TfR1) is largely unknown. We investigated whether iron availability affects TfR1 endocytic cycle and protein stability in HepG2 hepatoma cells exposed to ferric ammonium citrate (FAC). NH4Cl and bafilomycin A1, but not the proteasomal inhibitor MG132, prevented the FAC-mediated decrease in TfR1 protein levels, thus indicating lysosomal involvement. Knockdown experiments showed that TfR1 lysosomal degradation is independent of 1) endocytosis mediated by the clathrin adaptor AP2; 2) Tf, which was suggested to facilitate TfR1 internalization; 3) H-ferritin; and 4) MARCH8, previously implicated in TfR1 degradation. Notably, FAC decreased the number of TfR1 molecules at the cell surface and increased the Tf endocytic rate. Colocalization experiments confirmed that, upon FAC treatment, TfR1 was endocytosed in an AP2- and Tf-independent pathway and trafficked to the lysosome for degradation. This unconventional endocytic regulatory mechanism aimed at reducing surface TfR1 may represent an additional posttranslational control to prevent iron overload. Our results show that iron is a key regulator of the trafficking of TfR1, which has been widely used to study endocytosis, often not considering its function in iron homeostasis.
Collapse
Affiliation(s)
- Elena Gammella
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | | | - Alexia Conte
- IEO, Istituto Europeo di Oncologia IRCCS, 20141 Milan, Italy
| | - Stefano Freddi
- IEO, Istituto Europeo di Oncologia IRCCS, 20141 Milan, Italy
| | - Alessandra Alberghini
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Maura Poli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Sara Sigismund
- IEO, Istituto Europeo di Oncologia IRCCS, 20141 Milan, Italy.,Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, 20122, Milan, Italy
| | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Stefania Recalcati
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| |
Collapse
|