1
|
Mizuno K, Sugahara M, Kutomi O, Kato R, Itoh T, Fujita S, Yamada M. Direct observation of importin α family member KPNA1 in axonal transport with or without a schizophrenia-related mutation. J Biol Chem 2025; 301:108343. [PMID: 40010609 PMCID: PMC11982482 DOI: 10.1016/j.jbc.2025.108343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 02/03/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
Karyopherin α1 (KPNA1)/(human importin α5; mouse importin α1) facilitates cargo transport into the nucleus by forming a complex with a nuclear localization sequence containing cargo and importin β1 (IPOB1). The elevated KPNA1 expression in neurons and the correlation between mutations and psychiatric disorders suggest its broader significance beyond nucleocytoplasmic transport. Although KPNA1 is localized in the neurites of neurons, its role in axonal transport mechanisms remains unclear, and data on the connection between psychiatric disorders and signaling at the periphery of neurons remain limited. To address this knowledge gap, we investigated the dynamics of KPNA1 and related factors within axons. Our results showed that many of the axonal KPNA1 did not form a complex with IPOB1 in noninjured steady-state neurons. Axonal KPNA1 exhibited relatively stationary mobility and some showed bidirectional motility with fluctuating motion. KPNA1 partly comigrated with endosome/lysosome-associated factors, suggesting the presence of novel mechanisms underlie axonal transport and nucleocytoplasmic shuttling involving KPNA1 and IPOB1. Mutated KPNA1, which has been shown to be associated with psychiatric disorders (KPNA1E448X), was predominantly localized to the nucleus and lost from the axon. Incorporating a nuclear export signal (KPNA1E448X-NES) enhanced its subcellular localization and dynamics in the axon. Our findings demonstrate that KPNA1 functions not only as a shuttle between the cytoplasm and nucleus but also as a transporter in neuronal axons, relying on the endosomes for movement away from the nucleus with relatively slow net motions. Furthermore, a mutation in the Kpna1 gene can affect the dynamics of axonal transport. The insights from these mutations provide valuable knowledge for expanding our understanding of psychiatric disorders and facilitate the development of novel treatment strategies.
Collapse
Affiliation(s)
- Katsutoshi Mizuno
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui Prefecture, Japan; Life Science Innovation Center, University of Fukui, Fukui City, Fukui Prefecture, Japan
| | - Masaki Sugahara
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui Prefecture, Japan; Department of Frontier Fiber Technology and Science, Graduate School of Engineering, University of Fukui, Fukui City, Fukui Prefecture, Japan
| | - Osamu Kutomi
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui Prefecture, Japan; Life Science Innovation Center, University of Fukui, Fukui City, Fukui Prefecture, Japan
| | - Ryota Kato
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui Prefecture, Japan; Department of Frontier Fiber Technology and Science, Graduate School of Engineering, University of Fukui, Fukui City, Fukui Prefecture, Japan
| | - Takafumi Itoh
- Department of Bioscience and Biotechnology, Fukui Prefectural University, Yoshida-gun, Fukui Prefecture, Japan
| | - Satoshi Fujita
- Life Science Innovation Center, University of Fukui, Fukui City, Fukui Prefecture, Japan; Department of Frontier Fiber Technology and Science, Graduate School of Engineering, University of Fukui, Fukui City, Fukui Prefecture, Japan
| | - Masami Yamada
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui Prefecture, Japan; Life Science Innovation Center, University of Fukui, Fukui City, Fukui Prefecture, Japan.
| |
Collapse
|
2
|
Kodiha M, Azad N, Chu S, Crampton N, Stochaj U. Oxidative stress and signaling through EGFR and PKA pathways converge on the nuclear transport factor RanBP1. Eur J Cell Biol 2024; 103:151376. [PMID: 38011756 DOI: 10.1016/j.ejcb.2023.151376] [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: 09/30/2023] [Revised: 11/01/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
Nuclear protein trafficking requires the soluble transport factor RanBP1. The subcellular distribution of RanBP1 is dynamic, as the protein shuttles between the nucleus and cytoplasm. To date, the signaling pathways regulating RanBP1 subcellular localization are poorly understood. During interphase, RanBP1 resides mostly in the cytoplasm. We show here that oxidative stress concentrates RanBP1 in the nucleus, and our study defines the underlying mechanisms. Specifically, RanBP1's cysteine residues are not essential for its oxidant-induced relocation. Furthermore, our pharmacological approaches uncover that signaling mediated by epidermal growth factor receptor (EGFR) and protein kinase A (PKA) control RanBP1 localization during stress. In particular, pharmacological inhibitors of EGFR or PKA diminish the oxidant-dependent relocation of RanBP1. Mutant analysis identified serine 60 and tyrosine 103 as regulators of RanBP1 nuclear accumulation during oxidant exposure. Taken together, our results define RanBP1 as a target of oxidative stress and a downstream effector of EGFR and PKA signaling routes. This positions RanBP1 at the intersection of important cellular signaling circuits.
Collapse
Affiliation(s)
- Mohamed Kodiha
- Department of Physiology McGill University, Montreal H3G 1Y6, Canada
| | - Nabila Azad
- Department of Physiology McGill University, Montreal H3G 1Y6, Canada
| | - Siwei Chu
- Department of Physiology McGill University, Montreal H3G 1Y6, Canada
| | - Noah Crampton
- Department of Physiology McGill University, Montreal H3G 1Y6, Canada
| | - Ursula Stochaj
- Department of Physiology McGill University, Montreal H3G 1Y6, Canada.
| |
Collapse
|
3
|
Kumar R, Chhillar N, Gupta DS, Kaur G, Singhal S, Chauhan T. Cholesterol Homeostasis, Mechanisms of Molecular Pathways, and Cardiac Health: A Current Outlook. Curr Probl Cardiol 2024; 49:102081. [PMID: 37716543 DOI: 10.1016/j.cpcardiol.2023.102081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
The metabolism of lipoproteins, which regulate the transit of the lipid to and from tissues, is crucial to maintaining cholesterol homeostasis. Cardiac remodeling is referred to as a set of molecular, cellular, and interstitial changes that, following injury, affect the size, shape, function, mass, and geometry of the heart. Acetyl coenzyme A (acetyl CoA), which can be made from glucose, amino acids, or fatty acids, is the precursor for the synthesis of cholesterol. In this article, the authors explain concepts behind cardiac remodeling, its clinical ramifications, and the pathophysiological roles played by numerous various components, such as cell death, neurohormonal activation, oxidative stress, contractile proteins, energy metabolism, collagen, calcium transport, inflammation, and geometry. The levels of cholesterol are traditionally regulated by 2 biological mechanisms at the transcriptional stage. First, the SREBP transcription factor family regulates the transcription of crucial rate-limiting cholesterogenic and lipogenic proteins, which in turn limits cholesterol production. Immune cells become activated, differentiated, and divided, during an immune response with the objective of eradicating the danger signal. In addition to creating ATP, which is used as energy, this process relies on metabolic reprogramming of both catabolic and anabolic pathways to create metabolites that play a crucial role in regulating the response. Because of changes in signal transduction, malfunction of the sarcoplasmic reticulum and sarcolemma, impairment of calcium handling, increases in cardiac fibrosis, and progressive loss of cardiomyocytes, oxidative stress appears to be the primary mechanism that causes the transition from cardiac hypertrophy to heart failure. De novo cholesterol production, intestinal cholesterol absorption, and biliary cholesterol output are consequently crucial processes in cholesterol homeostasis. In the article's final section, the pharmacological management of cardiac remodeling is explored. The route of treatment is explained in different steps: including, promising, and potential strategies. This chapter offers a brief overview of the history of the study of cholesterol absorption as well as the different potential therapeutic targets.
Collapse
Affiliation(s)
| | - Neelam Chhillar
- Deparetment of Biochemistry, School of Medicine, DY Patil University, Navi Mumbai, India
| | - Dhruv Sanjay Gupta
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Ginpreet Kaur
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Shailey Singhal
- Cluster of Applied Sciences, University of Petroleum and Energy Studies, Dehradun, India
| | - Tanya Chauhan
- Division of Forensic Biology, National Forensic Sciences University, Delhi Campus (LNJN NICFS) Delhi, India
| |
Collapse
|
4
|
Shi J, Pei Y, Yu Q, Dong H. Progress in the study of parvovirus entry pathway. Virol J 2023; 20:61. [PMID: 37016419 PMCID: PMC10072039 DOI: 10.1186/s12985-023-02016-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/21/2023] [Indexed: 04/06/2023] Open
Abstract
A group of DNA viruses called parvoviruses that have significant effects on cancer therapy and genetic engineering applications. After passing through the cell membrane to reach the cytosol, it moves along the microtubule toward the nuclear membrane. The nuclear localization signal (NLS) is recognized by importin-beta (impβ) and other proteins from the complex outside the nuclear membrane and binds to enter the nucleus via the nuclear pore complex (NPC). There are two main pathways for viruses to enter the nucleus. The classical pathway is through the interaction of imp α and impβ with NLS via NPC. The other is the NPC mediated by the combination of impβ and it. While the capsid is introduced into the nucleus through classical nuclear transduction, there is also a transient nuclear membrane dissolution leading to passive transport into the nucleus, which has been proposed in recent years. This article mainly discusses several nuclear entry pathways and related proteins, providing a reference for subsequent research on viral entry pathways.
Collapse
Affiliation(s)
- Jiuming Shi
- College of Life Sciences, Jilin Agricultural University, Changchun, 130118, Jilin Province, China
| | - Yifeng Pei
- College of Life Sciences, Jilin Agricultural University, Changchun, 130118, Jilin Province, China
| | - Qian Yu
- College of Life Sciences, Jilin Agricultural University, Changchun, 130118, Jilin Province, China
| | - Hao Dong
- College of Life Sciences, Jilin Agricultural University, Changchun, 130118, Jilin Province, China.
- Engineering Research Center of Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
5
|
Audia S, Brescia C, Dattilo V, D’Antona L, Calvano P, Iuliano R, Trapasso F, Perrotti N, Amato R. RANBP1 (RAN Binding Protein 1): The Missing Genetic Piece in Cancer Pathophysiology and Other Complex Diseases. Cancers (Basel) 2023; 15:cancers15020486. [PMID: 36672435 PMCID: PMC9857238 DOI: 10.3390/cancers15020486] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
RANBP1 encoded by RANBP1 or HTF9A (Hpall Tiny Fragments Locus 9A), plays regulatory functions of the RAN-network, belonging to the RAS superfamily of small GTPases. Through this function, RANBP1 regulates the RANGAP1 activity and, thus, the fluctuations between GTP-RAN and GDP-RAN. In the light of this, RANBP1 take actions in maintaining the nucleus-cytoplasmic gradient, thus making nuclear import-export functional. RANBP1 has been implicated in the inter-nuclear transport of proteins, nucleic acids and microRNAs, fully contributing to cellular epigenomic signature. Recently, a RANBP1 diriment role in spindle checkpoint formation and nucleation has emerged, thus constituting an essential element in the control of mitotic stability. Over time, RANBP1 has been demonstrated to be variously involved in human cancers both for the role in controlling nuclear transport and RAN activity and for its ability to determine the efficiency of the mitotic process. RANBP1 also appears to be implicated in chemo-hormone and radio-resistance. A key role of this small-GTPases related protein has also been demonstrated in alterations of axonal flow and neuronal plasticity, as well as in viral and bacterial metabolism and in embryological maturation. In conclusion, RANBP1 appears not only to be an interesting factor in several pathological conditions but also a putative target of clinical interest.
Collapse
Affiliation(s)
- Salvatore Audia
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Carolina Brescia
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Vincenzo Dattilo
- Dipartimento di Medicina Sperimentale e Clinica, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Lucia D’Antona
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Pierluigi Calvano
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Rodolfo Iuliano
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Francesco Trapasso
- Dipartimento di Medicina Sperimentale e Clinica, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Nicola Perrotti
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Rosario Amato
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
- Correspondence: ; Tel.: +39-0961-3694084
| |
Collapse
|
6
|
Hu K, Yu H, Liu S, Liao D, Zhang Y. Systematic pan-cancer analysis on the expression and role of regulator of chromatin condensation 1/small nucleolar RNA host gene 3/small nucleolar RNA host gene 12. Front Mol Biosci 2022; 9:946507. [PMID: 36148010 PMCID: PMC9486007 DOI: 10.3389/fmolb.2022.946507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Regulator of chromatin condensation 1 (RCC1) is the major guanine nucleotide exchange factor of RAN GTPase, which plays a key role in various biological processes such as cell cycle and DNA damage repair. Small nucleolar RNA host gene 3 (SNHG3) and small nucleolar RNA host gene12 are long-stranded non-coding RNAs (lncRNAs) and are located on chromatin very close to the sequence of Regulator of chromatin condensation 1. Many studies have shown that they are aberrantly expressed in tumor tissues and can affect the proliferation and viability of cancer cells. Although the effects of Regulator of chromatin condensation 1/small nucleolar RNA host gene 3/small nucleolar RNA host gene12 on cellular activity have been reported, respectively, their overall analysis on the pan-cancer level has not been performed. Here, we performed a comprehensive analysis of Regulator of chromatin condensation 1/small nucleolar RNA host gene 3/small nucleolar RNA host gene12 in 33 cancers through the Cancer Genome Atlas and Gene Expression Database. The results showed that Regulator of chromatin condensation 1/small nucleolar RNA host gene 3/small nucleolar RNA host gene12 were highly expressed in a variety of tumor tissues compared to normal tissues. The expression of Regulator of chromatin condensation 1/small nucleolar RNA host gene 3/small nucleolar RNA host gene12 in BRCA, LGG and LIHC was associated with TP53 mutations. In addition, Regulator of chromatin condensation 1/small nucleolar RNA host gene 3/small nucleolar RNA host gene12 expression was closely associated with the prognosis of patients with multiple tumors. Immunocorrelation analysis indicated that Regulator of chromatin condensation 1/small nucleolar RNA host gene 3/small nucleolar RNA host gene12 showed a correlation with multiple immune cell infiltration. The results of enrichment analysis suggested that Regulator of chromatin condensation 1/small nucleolar RNA host gene 3/small nucleolar RNA host gene12 was involved in the regulation of cell cycle, apoptosis and other pathways. We found that these effects were mainly mediated by Regulator of chromatin condensation 1, while the trend of small nucleolar RNA host gene 3/small nucleolar RNA host gene12 regulation was also consistent with regulator of chromatin condensation 1. The important role played by Regulator of chromatin condensation 1 in tumor diseases was further corroborated by the study of adjacent lncRNAs.These findings provide new and comprehensive insights into the role of Regulator of chromatin condensation 1/small nucleolar RNA host gene 3/small nucleolar RNA host gene12 in tumor development and show their potential as clinical monitoring and therapy.
Collapse
|
7
|
Li H, Ho LWC, Lee LKC, Liu S, Chan CKW, Tian XY, Choi CHJ. Intranuclear Delivery of DNA Nanostructures via Cellular Mechanotransduction. NANO LETTERS 2022; 22:3400-3409. [PMID: 35436127 DOI: 10.1021/acs.nanolett.2c00667] [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] [Indexed: 06/14/2023]
Abstract
DNA nanostructures are attractive gene carriers for nanomedicine applications, yet their delivery to the nucleus remains inefficient. We present the application of extracellular mechanical stimuli to activate cellular mechanotransduction for boosting the intranuclear delivery of DNA nanostructures. Treating mammalian cells with polythymidine-rich spherical nucleic acids (poly(T) SNAs) under gentle compression by a single coverslip leads to up to ∼50% nuclear accumulation without severe endosomal entrapment, cytotoxicity, or long-term membrane damage; no chemical modification or transfection reagent is needed. Gentle compression activates Rho-ROCK mechanotransduction and causes nuclear translocation of YAP. Joint compression and treatment with poly(T) oligonucleotides upregulate genes linked to myosin, actin filament, and nuclear import. In turn, Rho-ROCK, myosin, and importin mediate the nuclear entry of poly(T) SNAs. Treatment of endothelioma cells with poly(T) SNAs bearing antisense oligonucleotides under compression inhibits an intranuclear oncogene. Our data should inspire the marriage of DNA nanotechnology and cellular biomechanics for intranuclear applications.
Collapse
|
8
|
Merlo LM, Peng W, DuHadaway JB, Montgomery JD, Prendergast GC, Muller AJ, Mandik-Nayak L. The Immunomodulatory Enzyme IDO2 Mediates Autoimmune Arthritis through a Nonenzymatic Mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:571-581. [PMID: 34965962 PMCID: PMC8770583 DOI: 10.4049/jimmunol.2100705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/15/2021] [Indexed: 02/03/2023]
Abstract
IDO2 is one of two closely related tryptophan catabolizing enzymes induced under inflammatory conditions. In contrast to the immunoregulatory role defined for IDO1 in cancer models, IDO2 has a proinflammatory function in models of autoimmunity and contact hypersensitivity. In humans, two common single-nucleotide polymorphisms have been identified that severely impair IDO2 enzymatic function, such that <25% of individuals express IDO2 with full catalytic potential. This, together with IDO2's relatively weak enzymatic activity, suggests that IDO2 may have a role outside of its function in tryptophan catabolism. To determine whether the enzymatic activity of IDO2 is required for its proinflammatory function, we used newly generated catalytically inactive IDO2 knock-in mice together with established models of contact hypersensitivity and autoimmune arthritis. Contact hypersensitivity was attenuated in catalytically inactive IDO2 knock-in mice. In contrast, induction of autoimmune arthritis was unaffected by the absence of IDO2 enzymatic activity. In pursuing this nonenzymatic IDO2 function, we identified GAPDH, Runx1, RANbp10, and Mgea5 as IDO2-binding proteins that do not interact with IDO1, implicating them as potential mediators of IDO2-specific function. Taken together, our findings identify a novel function for IDO2, independent of its tryptophan catabolizing activity, and suggest that this nonenzymatic function could involve multiple signaling pathways. These data show that the enzymatic activity of IDO2 is required only for some inflammatory immune responses and provide, to our knowledge, the first evidence of a nonenzymatic role for IDO2 in mediating autoimmune disease.
Collapse
Affiliation(s)
| | - Weidan Peng
- Lankenau Institute for Medical Research, Wynnewood, PA
| | | | | | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA,Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | | |
Collapse
|
9
|
Tao M, Xiao K, Zheng Y, Li Z, Luo Q, Wang G, Hu Z. Identification and characterization of a novel Channelrhodopsin gene HpChR1 in Haematococcus pluvialis. ALGAL RES 2021. [DOI: 10.1016/j.algal.2021.102263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
10
|
Böhnke J, Pinkert S, Schmidt M, Binder H, Bilz NC, Jung M, Reibetanz U, Beling A, Rujescu D, Claus C. Coxsackievirus B3 Infection of Human iPSC Lines and Derived Primary Germ-Layer Cells Regarding Receptor Expression. Int J Mol Sci 2021; 22:1220. [PMID: 33513663 PMCID: PMC7865966 DOI: 10.3390/ijms22031220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 02/06/2023] Open
Abstract
The association of members of the enterovirus family with pregnancy complications up to miscarriages is under discussion. Here, infection of two different human induced pluripotent stem cell (iPSC) lines and iPSC-derived primary germ-layer cells with coxsackievirus B3 (CVB3) was characterized as an in vitro cell culture model for very early human development. Transcriptomic analysis of iPSC lines infected with recombinant CVB3 expressing enhanced green fluorescent protein (EGFP) revealed a reduction in the expression of pluripotency genes besides an enhancement of genes involved in RNA metabolism. The initial distribution of CVB3-EGFP-positive cells within iPSC colonies correlated with the distribution of its receptor coxsackie- and adenovirus receptor (CAR). Application of anti-CAR blocking antibodies supported the requirement of CAR, but not of the co-receptor decay-accelerating factor (DAF) for infection of iPSC lines. Among iPSC-derived germ-layer cells, mesodermal cells were especially vulnerable to CVB3-EGFP infection. Our data implicate further consideration of members of the enterovirus family in the screening program of human pregnancies. Furthermore, iPSCs with their differentiation capacity into cell populations of relevant viral target organs could offer a reliable screening approach for therapeutic intervention and for assessment of organ-specific enterovirus virulence.
Collapse
Affiliation(s)
- Janik Böhnke
- Institute of Medical Microbiology and Virology, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany; (J.B.); (N.C.B.)
| | - Sandra Pinkert
- Institute of Biochemistry, Berlin Institute of Health (BIH) and Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (S.P.); (A.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Side, 10115 Berlin, Germany
| | - Maria Schmidt
- Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107 Leipzig, Germany; (M.S.); (H.B.)
| | - Hans Binder
- Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107 Leipzig, Germany; (M.S.); (H.B.)
| | - Nicole Christin Bilz
- Institute of Medical Microbiology and Virology, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany; (J.B.); (N.C.B.)
| | - Matthias Jung
- Department of Psychiatry, Psychotherapy, and Psychosomatic Medicine, Martin Luther University Halle Wittenberg, Julius-Kuehn-Strasse 7, 06112 Halle (Saale), Germany; (M.J.); (D.R.)
| | - Uta Reibetanz
- Institute for Medical Physics and Biophysics, Medical Faculty, University of Leipzig, Härtelstrasse 16-18, 04107 Leipzig, Germany;
| | - Antje Beling
- Institute of Biochemistry, Berlin Institute of Health (BIH) and Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (S.P.); (A.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Side, 10115 Berlin, Germany
| | - Dan Rujescu
- Department of Psychiatry, Psychotherapy, and Psychosomatic Medicine, Martin Luther University Halle Wittenberg, Julius-Kuehn-Strasse 7, 06112 Halle (Saale), Germany; (M.J.); (D.R.)
| | - Claudia Claus
- Institute of Medical Microbiology and Virology, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany; (J.B.); (N.C.B.)
| |
Collapse
|
11
|
Zhou J, Tan Y, Zhang Y, Tong A, Shen X, Sun X, Jia D, Sun Q. GEF-independent Ran activation shifts a fraction of the protein to the cytoplasm and promotes cell proliferation. MOLECULAR BIOMEDICINE 2020; 1:18. [PMID: 35006421 PMCID: PMC8607414 DOI: 10.1186/s43556-020-00011-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
Ran (Ras-related nuclear protein) plays several important roles in nucleo-cytoplasmic transport, mitotic spindle formation, nuclear envelope/nuclear pore complex assembly, and other functions in the cytoplasm, as well as in cellular transformation when switched on. Unlike other members of the GTPase superfamily, Ran binds more tightly to GDP than to GTP due to the presence of an auto-inhibitory C-terminal tail. Multiple missense mutations in the C-terminus of Ran occur in cancers, but their biological significance remains unclear. Here, the quantitative GDP/GTP binding preference of four engineered mutations with unstable C-termini was analyzed using a devised mant-GDP dissociation assay. The results showed that the impact of different C-terminal mutations depends on multiple factors. Although these mutants were more GTP-loaded in human cells, they were shown to be more cytoplasmic, and to support nuclear transport with minimally or partially reduced efficiency. Further, several Ran cancer mutants were compromised in autoinhibition, slightly more GTP-bound, more cytoplasmic, and enhanced the proliferation of A549 and HeLa cells in vitro. Thus, our work reveals a new route of Ran activation independent of guanine nucleotide exchange factor (GEF), which may account for the hyper-proliferation induced by Ran cancer mutations.
Collapse
Affiliation(s)
- Jinhan Zhou
- Department of Pathology, State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Yuping Tan
- Department of Pathology, State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Yuqing Zhang
- Department of Pathology, State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Aiping Tong
- Department of Pathology, State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Xiaofei Shen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, Division of Neurology, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaodong Sun
- Department of Pharmacology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, Division of Neurology, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qingxiang Sun
- Department of Pathology, State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
12
|
Sahoo PK, Kar AN, Samra N, Terenzio M, Patel P, Lee SJ, Miller S, Thames E, Jones B, Kawaguchi R, Coppola G, Fainzilber M, Twiss JL. A Ca 2+-Dependent Switch Activates Axonal Casein Kinase 2α Translation and Drives G3BP1 Granule Disassembly for Axon Regeneration. Curr Biol 2020; 30:4882-4895.e6. [PMID: 33065005 DOI: 10.1016/j.cub.2020.09.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 07/15/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
The main limitation on axon regeneration in the peripheral nervous system (PNS) is the slow rate of regrowth. We recently reported that nerve regeneration can be accelerated by axonal G3BP1 granule disassembly, releasing axonal mRNAs for local translation to support axon growth. Here, we show that G3BP1 phosphorylation by casein kinase 2α (CK2α) triggers G3BP1 granule disassembly in injured axons. CK2α activity is temporally and spatially regulated by local translation of Csnk2a1 mRNA in axons after injury, but this requires local translation of mTor mRNA and buffering of the elevated axonal Ca2+ that occurs after axotomy. CK2α's appearance in axons after PNS nerve injury correlates with disassembly of axonal G3BP1 granules as well as increased phospho-G3BP1 and axon growth, although depletion of Csnk2a1 mRNA from PNS axons decreases regeneration and increases G3BP1 granules. Phosphomimetic G3BP1 shows remarkably decreased RNA binding in dorsal root ganglion (DRG) neurons compared with wild-type and non-phosphorylatable G3BP1; combined with other studies, this suggests that CK2α-dependent G3BP1 phosphorylation on Ser 149 after axotomy releases axonal mRNAs for translation. Translation of axonal mRNAs encoding some injury-associated proteins is known to be increased with Ca2+ elevations, and using a dual fluorescence recovery after photobleaching (FRAP) reporter assay for axonal translation, we see that translational specificity switches from injury-associated protein mRNA translation to CK2α translation with endoplasmic reticulum (ER) Ca2+ release versus cytoplasmic Ca2+ chelation. Our results point to axoplasmic Ca2+ concentrations as a determinant for the temporal specificity of sequential translational activation of different axonal mRNAs as severed axons transition from injury to regenerative growth.
Collapse
Affiliation(s)
- Pabitra K Sahoo
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Amar N Kar
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Nitzan Samra
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovat, Israel
| | - Marco Terenzio
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovat, Israel; Molecular Neuroscience Unit, Okinawa Institute of Science and Technology, Kunigami, Okinawa 904-0412, Japan
| | - Priyanka Patel
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Seung Joon Lee
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Sharmina Miller
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Elizabeth Thames
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Blake Jones
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Riki Kawaguchi
- Department of Neurology, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA 90095-1761, USA
| | - Giovanni Coppola
- Department of Neurology, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA 90095-1761, USA
| | - Mike Fainzilber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovat, Israel
| | - Jeffery L Twiss
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
13
|
Swart AL, Gomez-Valero L, Buchrieser C, Hilbi H. Evolution and function of bacterial RCC1 repeat effectors. Cell Microbiol 2020; 22:e13246. [PMID: 32720355 DOI: 10.1111/cmi.13246] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 01/02/2023]
Abstract
Intracellular bacterial pathogens harbour genes, the closest homologues of which are found in eukaryotes. Regulator of chromosome condensation 1 (RCC1) repeat proteins are phylogenetically widespread and implicated in protein-protein interactions, such as the activation of the small GTPase Ran by its cognate guanine nucleotide exchange factor, RCC1. Legionella pneumophila and Coxiella burnetii, the causative agents of Legionnaires' disease and Q fever, respectively, harbour RCC1 repeat coding genes. Legionella pneumophila secretes the RCC1 repeat 'effector' proteins LegG1, PpgA and PieG into eukaryotic host cells, where they promote the activation of the pleiotropic small GTPase Ran, microtubule stabilisation, pathogen vacuole motility and intracellular bacterial growth as well as host cell migration. The RCC1 repeat effectors localise to the pathogen vacuole or the host plasma membrane and target distinct components of the Ran GTPase cycle, including Ran modulators and the small GTPase itself. Coxiella burnetii translocates the RCC1 repeat effector NopA into host cells, where the protein localises to nucleoli. NopA binds to Ran GTPase and promotes the nuclear accumulation of Ran(GTP), thus pertubing the import of the transcription factor NF-κB and innate immune signalling. Hence, divergent evolution of bacterial RCC1 repeat effectors defines the range of Ran GTPase cycle targets and likely allows fine-tuning of Ran GTPase activation by the pathogens at different cellular sites.
Collapse
Affiliation(s)
- Anna Leoni Swart
- Institute of Medical Microbiology, Faculty of Medicine, University of Zurich, Zürich, Switzerland
| | - Laura Gomez-Valero
- Institut Pasteur, Unité de Biologie des Bactéries Intracellulaires, Paris, France.,CNRS UMR 3525, Paris, France
| | - Carmen Buchrieser
- Institut Pasteur, Unité de Biologie des Bactéries Intracellulaires, Paris, France.,CNRS UMR 3525, Paris, France
| | - Hubert Hilbi
- Institute of Medical Microbiology, Faculty of Medicine, University of Zurich, Zürich, Switzerland
| |
Collapse
|
14
|
Divergent Evolution of Legionella RCC1 Repeat Effectors Defines the Range of Ran GTPase Cycle Targets. mBio 2020; 11:mBio.00405-20. [PMID: 32209684 PMCID: PMC7157520 DOI: 10.1128/mbio.00405-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Legionella pneumophila is a ubiquitous environmental bacterium which, upon inhalation, causes a life-threatening pneumonia termed Legionnaires’ disease. The opportunistic pathogen grows in amoebae and macrophages by employing a “type IV” secretion system, which secretes more than 300 different “effector” proteins into the host cell, where they subvert pivotal processes. The function of many of these effector proteins is unknown, and their evolution has not been studied. L. pneumophila RCC1 repeat effectors target the small GTPase Ran, a molecular switch implicated in different cellular processes such as nucleocytoplasmic transport and microtubule cytoskeleton dynamics. We provide evidence that one or more RCC1 repeat genes are distributed in two main clusters of L. pneumophila strains and have divergently evolved to target different components of the Ran GTPase activation cycle at different subcellular sites. Thus, L. pneumophila employs a sophisticated strategy to subvert host cell Ran GTPase during infection. Legionella pneumophila governs its interactions with host cells by secreting >300 different “effector” proteins. Some of these effectors contain eukaryotic domains such as the RCC1 (regulator of chromosome condensation 1) repeats promoting the activation of the small GTPase Ran. In this report, we reveal a conserved pattern of L. pneumophila RCC1 repeat genes, which are distributed in two main clusters of strains. Accordingly, strain Philadelphia-1 contains two RCC1 genes implicated in bacterial virulence, legG1 (Legionella eukaryotic gene 1), and ppgA, while strain Paris contains only one, pieG. The RCC1 repeat effectors localize to different cellular compartments and bind distinct components of the Ran GTPase cycle, including Ran modulators and the small GTPase itself, and yet they all promote the activation of Ran. The pieG gene spans the corresponding open reading frames of legG1 and a separate adjacent upstream gene, lpg1975. legG1 and lpg1975 are fused upon addition of a single nucleotide to encode a protein that adopts the binding specificity of PieG. Thus, a point mutation in pieG splits the gene, altering the effector target. These results indicate that divergent evolution of RCC1 repeat effectors defines the Ran GTPase cycle targets and that modulation of different components of the cycle might fine-tune Ran activation during Legionella infection.
Collapse
|
15
|
Ran promotes membrane targeting and stabilization of RhoA to orchestrate ovarian cancer cell invasion. Nat Commun 2019; 10:2666. [PMID: 31209254 PMCID: PMC6573066 DOI: 10.1038/s41467-019-10570-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 05/15/2019] [Indexed: 12/22/2022] Open
Abstract
Ran is a nucleocytoplasmic shuttle protein that is involved in cell cycle regulation, nuclear-cytoplasmic transport, and cell transformation. Ran plays an important role in cancer cell survival and cancer progression. Here, we show that, in addition to the nucleocytoplasmic localization of Ran, this GTPase is specifically associated with the plasma membrane/ruffles of ovarian cancer cells. Ran depletion has a drastic effect on RhoA stability and inhibits RhoA localization to the plasma membrane/ruffles and RhoA activity. We further demonstrate that the DEDDDL domain of Ran is required for the interaction with serine 188 of RhoA, which prevents RhoA degradation by the proteasome pathway. Moreover, the knockdown of Ran leads to a reduction of ovarian cancer cell invasion by impairing RhoA signalling. Our findings provide advanced insights into the mode of action of the Ran-RhoA signalling axis and may represent a potential therapeutic avenue for drug development to prevent ovarian tumour metastasis. Ran, a nucleus-cytoplasm shuttle protein, is implicated in cancer development and survival. Here, the authors show that Ran binds RhoA to impair its degradation and allow its localisation to the plasma membrane of ovarian cancer cells for tumour invasion.
Collapse
|
16
|
Liang HX, Liu HW. Inducible Expression of Ran1 and Its GDP- and GTP-Bound Mimetic Mutants Leads to Defects in Amitosis and Cytokinesis with Abnormal Cytoplasmic Microtubule Assembly. Mol Biol 2019. [DOI: 10.1134/s0026893319030105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
17
|
Prieto-Dominguez N, Parnell C, Teng Y. Drugging the Small GTPase Pathways in Cancer Treatment: Promises and Challenges. Cells 2019; 8:E255. [PMID: 30884855 PMCID: PMC6468615 DOI: 10.3390/cells8030255] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023] Open
Abstract
Small GTPases are a family of low molecular weight GTP-hydrolyzing enzymes that cycle between an inactive state when bound to GDP and an active state when associated to GTP. Small GTPases regulate key cellular processes (e.g., cell differentiation, proliferation, and motility) as well as subcellular events (e.g., vesicle trafficking), making them key participants in a great array of pathophysiological processes. Indeed, the dysfunction and deregulation of certain small GTPases, such as the members of the Ras and Arf subfamilies, have been related with the promotion and progression of cancer. Therefore, the development of inhibitors that target dysfunctional small GTPases could represent a potential therapeutic strategy for cancer treatment. This review covers the basic biochemical mechanisms and the diverse functions of small GTPases in cancer. We also discuss the strategies and challenges of inhibiting the activity of these enzymes and delve into new approaches that offer opportunities to target them in cancer therapy.
Collapse
Affiliation(s)
- Néstor Prieto-Dominguez
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Institute of Biomedicine (IBIOMED), University of León, León 24010, Spain.
| | | | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Department of Medical laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
18
|
Drosophila nucleostemin 3 is required to maintain larval neuroblast proliferation. Dev Biol 2018; 440:1-12. [PMID: 29679561 PMCID: PMC6278609 DOI: 10.1016/j.ydbio.2018.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/14/2018] [Accepted: 04/17/2018] [Indexed: 02/03/2023]
Abstract
Stem cells must maintain proliferation during tissue development, repair and homeostasis, yet avoid tumor formation. In Drosophila, neural stem cells (neuroblasts) maintain proliferation during embryonic and larval development and terminate cell cycle during metamorphosis. An important question for understanding how tissues are generated and maintained is: what regulates stem cell proliferation versus differentiation? We performed a genetic screen which identified nucleostemin 3 (ns3) as a gene required to maintain neuroblast proliferation. ns3 is evolutionarily conserved with yeast and human Lsg1, which encode putative GTPases and are essential for organism growth and viability. We found NS3 is cytoplasmic and it is required to retain the cell cycle repressor Prospero in neuroblast cytoplasm via a Ran-independent pathway. NS3 is also required for proper neuroblast cell polarity and asymmetric cell division. Structure-function analysis further shows that the GTP-binding domain and acidic domain are required for NS3 function in neuroblast proliferation. We conclude NS3 has novel roles in regulating neuroblast cell polarity and proliferation.
Collapse
|
19
|
Sharma A, Tiwari M, Gupta A, Pandey AN, Yadav PK, Chaube SK. Journey of oocyte from metaphase-I to metaphase-II stage in mammals. J Cell Physiol 2018; 233:5530-5536. [PMID: 29331044 DOI: 10.1002/jcp.26467] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/05/2018] [Indexed: 12/13/2022]
Abstract
In mammals, journey from metaphase-I (M-I) to metaphase-II (M-II) is important since oocyte extrude first polar body (PB-I) and gets converted into haploid gamete. The molecular and cellular changes associated with meiotic cell cycle progression from M-I to M-II stage and extrusion of PB-I remain ill understood. Several factors drive oocyte meiosis from M-I to M-II stage. The mitogen-activated protein kinase3/1 (MAPK3/1), signal molecules and Rho family GTPases act through various pathways to drive cell cycle progression from M-I to M-II stage. The down regulation of MOS/MEK/MAPK3/1 pathway results in the activation of anaphase-promoting complex/cyclosome (APC/C). The active APC/C destabilizes maturation promoting factor (MPF) and induces meiotic resumption. Several signal molecules such as, c-Jun N-terminal kinase (JNK2), SENP3, mitotic kinesin-like protein 2 (MKlp2), regulator of G-protein signaling (RGS2), Epsin2, polo-like kinase 1 (Plk1) are directly or indirectly involved in chromosomal segregation. Rho family GTPase is another enzyme that along with cell division cycle (Cdc42) to form actomyosin contractile ring required for chromosomal segregation. In the presence of origin recognition complex (ORC4), eccentrically localized haploid set of chromosomes trigger cortex differentiation and determine the division site for polar body formation. The actomyosin contractile activity at the site of division plane helps to form cytokinetic furrow that results in the formation and extrusion of PB-I. Indeed, oocyte journey from M-I to M-II stage is coordinated by several factors and pathways that enable oocyte to extrude PB-I. Quality of oocyte directly impact fertilization rate, early embryonic development, and reproductive outcome in mammals.
Collapse
Affiliation(s)
- Alka Sharma
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Meenakshi Tiwari
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Anumegha Gupta
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashutosh N Pandey
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Pramod K Yadav
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Shail K Chaube
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
20
|
Swart AL, Harrison CF, Eichinger L, Steinert M, Hilbi H. Acanthamoeba and Dictyostelium as Cellular Models for Legionella Infection. Front Cell Infect Microbiol 2018; 8:61. [PMID: 29552544 PMCID: PMC5840211 DOI: 10.3389/fcimb.2018.00061] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/13/2018] [Indexed: 12/20/2022] Open
Abstract
Environmental bacteria of the genus Legionella naturally parasitize free-living amoebae. Upon inhalation of bacteria-laden aerosols, the opportunistic pathogens grow intracellularly in alveolar macrophages and can cause a life-threatening pneumonia termed Legionnaires' disease. Intracellular replication in amoebae and macrophages takes place in a unique membrane-bound compartment, the Legionella-containing vacuole (LCV). LCV formation requires the bacterial Icm/Dot type IV secretion system, which translocates literally hundreds of "effector" proteins into host cells, where they modulate crucial cellular processes for the pathogen's benefit. The mechanism of LCV formation appears to be evolutionarily conserved, and therefore, amoebae are not only ecologically significant niches for Legionella spp., but also useful cellular models for eukaryotic phagocytes. In particular, Acanthamoeba castellanii and Dictyostelium discoideum emerged over the last years as versatile and powerful models. Using genetic, biochemical and cell biological approaches, molecular interactions between amoebae and Legionella pneumophila have recently been investigated in detail with a focus on the role of phosphoinositide lipids, small and large GTPases, autophagy components and the retromer complex, as well as on bacterial effectors targeting these host factors.
Collapse
Affiliation(s)
- A Leoni Swart
- Institute of Medical Microbiology, Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Christopher F Harrison
- Max von Pettenkofer Institute, Medical Faculty, Ludwig-Maximilians University Munich, Munich, Germany
| | - Ludwig Eichinger
- Institute for Biochemistry I, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Michael Steinert
- Department of Life Sciences, Institute of Microbiology, Technical University of Braunschweig, Braunschweig, Germany
| | - Hubert Hilbi
- Institute of Medical Microbiology, Medical Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Puverel S, Kiris E, Singh S, Klarmann KD, Coppola V, Keller JR, Tessarollo L. RanBPM (RanBP9) regulates mouse c-Kit receptor level and is essential for normal development of bone marrow progenitor cells. Oncotarget 2018; 7:85109-85123. [PMID: 27835883 PMCID: PMC5341297 DOI: 10.18632/oncotarget.13198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 10/26/2016] [Indexed: 01/22/2023] Open
Abstract
c-Kit is a tyrosine kinase receptor important for gametogenesis, hematopoiesis, melanogenesis and mast cell biology. Dysregulation of c-Kit function is oncogenic and its expression in the stem cell niche of a number of tissues has underlined its relevance for regenerative medicine and hematopoietic stem cell biology. Yet, very little is known about the mechanisms that control c-Kit protein levels. Here we show that the RanBPM/RanBP9 scaffold protein binds to c-Kit and is necessary for normal c-Kit protein expression in the mouse testis and subset lineages of the hematopoietic system. RanBPM deletion causes a reduction in c-Kit protein but not its mRNA suggesting a posttranslational mechanism. This regulation is specific to the c-Kit receptor since RanBPM reduction does not affect other membrane proteins examined. Importantly, in both mouse hematopoietic system and testis, RanBPM deficiency causes defects consistent with c-Kit loss of expression suggesting that RanBPM is an important regulator of c-Kit function. The finding that this regulatory mechanism is also present in human cells expressing endogenous RanBPM and c-Kit suggests a potential new strategy to target oncogenic c-Kit in malignancies.
Collapse
Affiliation(s)
- Sandrine Puverel
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Erkan Kiris
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Satyendra Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Kimberly D Klarmann
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA.,Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Vincenzo Coppola
- The Ohio State University, Department of Cancer, Biology and Genetics, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Jonathan R Keller
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA.,Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| |
Collapse
|
22
|
Wang MJ, Xu YY, Huang RY, Chen XM, Chen HM, Han L, Yan YH, Lu CJ. Role of an imbalanced miRNAs axis in pathogenesis of psoriasis: novel perspectives based on review of the literature. Oncotarget 2018; 8:5498-5507. [PMID: 27729619 PMCID: PMC5354926 DOI: 10.18632/oncotarget.12534] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/03/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Specific profile of microRNAs (miRNAs, miR) expressed in psoriasis has been identified in the past few years, while the studies on roles and molecular mechanisms of these miRNAs are still on the way. In our previous study, four specific miRNAs (miR-31, miR-203, hsa-miR-99a and miR-125b) were found to be specifically altered in psoriatic lesions.We therefore conducted a systematic literature review in this study to reveal the role of these miRNAs in the pathogenesis of psoriasis in order to inform future research. METHODS The related articles indexed in PubMed (MEDLINE) database were searched and analyzed. We identified eligible studies related to the mechanism research of miR-31, miR-203, hsa-miR-99a and miR-125b in psoriasis or psoriatic lesional skin from inception up to July 2016. The experts in the field of miRNAs and Psoriasis were involved in analysis process. RESULT Both miR-31 and miR-203 are dramatically upregulated in psoriatic lesions. The former plays the pro-proliferative, pro-differentiative and pro-inflammatory roles and the latter holds the potentials for anti-proliferation, pro-inflammation and pro-differentiation in psoriatic keratinocytes. Conversely, both hsa-miR-99a and miR-125b are significantly downregulated in psoriatic skin. These two miRNAs are able to inhibit proliferation while promote differentiation of psoriatic keratinocytes, and miR-125b can also suppress inflammation in psoriatic lesions. By analyzing the contexts related to these miRNAs, we found that each of them does not act alone but rather work in concert with other miRNAs. The imbalance between miR-31/miR-203and hsa-miR-99a/miR-125b may contribute to the intense proliferation and abnormal differentiation of psoriatic keratinocytes, which is a characteristic of pathogenesis of psoriasis. CONCLUSION An imbalanced miRNAs axis was for the first time outlined. Apparently, upregulation of miR-31/miR-203 and downregulation of hsa-miR-99a/miR-125b work together in concert to facilitate the development of psoriasis pathogenesis. Further work in this field holds the potentials to open a new way to study psoriasis.
Collapse
Affiliation(s)
- Mao-Jie Wang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Yong-Yue Xu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Run-Yue Huang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Section of Metabolic Diseases Research, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Xiu-Min Chen
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Hai-Ming Chen
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Ling Han
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Yu-Hong Yan
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Chuan-Jian Lu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| |
Collapse
|
23
|
Maffioli E, Schulte C, Nonnis S, Grassi Scalvini F, Piazzoni C, Lenardi C, Negri A, Milani P, Tedeschi G. Proteomic Dissection of Nanotopography-Sensitive Mechanotransductive Signaling Hubs that Foster Neuronal Differentiation in PC12 Cells. Front Cell Neurosci 2018; 11:417. [PMID: 29354032 PMCID: PMC5758595 DOI: 10.3389/fncel.2017.00417] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022] Open
Abstract
Neuronal cells are competent in precisely sensing nanotopographical features of their microenvironment. The perceived microenvironmental information will be “interpreted” by mechanotransductive processes and impacts on neuronal functioning and differentiation. Attempts to influence neuronal differentiation by engineering substrates that mimic appropriate extracellular matrix (ECM) topographies are hampered by the fact that profound details of mechanosensing/-transduction complexity remain elusive. Introducing omics methods into these biomaterial approaches has the potential to provide a deeper insight into the molecular processes and signaling cascades underlying mechanosensing/-transduction but their exigence in cellular material is often opposed by technical limitations of major substrate top-down fabrication methods. Supersonic cluster beam deposition (SCBD) allows instead the bottom-up fabrication of nanostructured substrates over large areas characterized by a quantitatively controllable ECM-like nanoroughness that has been recently shown to foster neuron differentiation and maturation. Exploiting this capacity of SCBD, we challenged mechanosensing/-transduction and differentiative behavior of neuron-like PC12 cells with diverse nanotopographies and/or changes of their biomechanical status, and analyzed their phosphoproteomic profiles in these settings. Versatile proteins that can be associated to significant processes along the mechanotransductive signal sequence, i.e., cell/cell interaction, glycocalyx and ECM, membrane/f-actin linkage and integrin activation, cell/substrate interaction, integrin adhesion complex, actomyosin organization/cellular mechanics, nuclear organization, and transcriptional regulation, were affected. The phosphoproteomic data suggested furthermore an involvement of ILK, mTOR, Wnt, and calcium signaling in these nanotopography- and/or cell mechanics-related processes. Altogether, potential nanotopography-sensitive mechanotransductive signaling hubs participating in neuronal differentiation were dissected.
Collapse
Affiliation(s)
- Elisa Maffioli
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Carsten Schulte
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Simona Nonnis
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Claudio Piazzoni
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Cristina Lenardi
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Armando Negri
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Paolo Milani
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| |
Collapse
|
24
|
Aramburu IV, Lemke EA. Floppy but not sloppy: Interaction mechanism of FG-nucleoporins and nuclear transport receptors. Semin Cell Dev Biol 2017; 68:34-41. [PMID: 28669824 PMCID: PMC7611744 DOI: 10.1016/j.semcdb.2017.06.026] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 06/29/2017] [Indexed: 12/15/2022]
Abstract
The nuclear pore complex (NPC) forms a permeability barrier between the nucleus and the cytoplasm. Molecules that are able to cross this permeability barrier encounter different disordered phenylalanine glycine rich nucleoporins (FG-Nups) that act as a molecular filter and regulate the selective NPC crossing of biomolecules. In this review, we provide a current overview regarding the interaction mechanism between FG-Nups and the carrier molecules that recognize and enable the transport of cargoes through the NPC aiming to understand the general molecular mechanisms that facilitate the nucleocytoplasmic transport.
Collapse
Affiliation(s)
- Iker Valle Aramburu
- Structural and Computational Biology Unit and Cell Biology and Biophysics Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Edward A Lemke
- Structural and Computational Biology Unit and Cell Biology and Biophysics Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany.
| |
Collapse
|
25
|
Ran-dependent TPX2 activation promotes acentrosomal microtubule nucleation in neurons. Sci Rep 2017; 7:42297. [PMID: 28205572 PMCID: PMC5304320 DOI: 10.1038/srep42297] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/05/2017] [Indexed: 01/07/2023] Open
Abstract
The microtubule (MT) cytoskeleton is essential for the formation of morphologically appropriate neurons. The existence of the acentrosomal MT organizing center in neurons has been proposed but its identity remained elusive. Here we provide evidence showing that TPX2 is an important component of this acentrosomal MT organizing center. First, neurite elongation is compromised in TPX2-depleted neurons. In addition, TPX2 localizes to the centrosome and along the neurite shaft bound to MTs. Depleting TPX2 decreases MT formation frequency specifically at the tip and the base of the neurite, and these correlate precisely with the regions where active GTP-bound Ran proteins are enriched. Furthermore, overexpressing the downstream effector of Ran, importin, compromises MT formation and neuronal morphogenesis. Finally, applying a Ran-importin signaling interfering compound phenocopies the effect of TPX2 depletion on MT dynamics. Together, these data suggest a model in which Ran-dependent TPX2 activation promotes acentrosomal MT nucleation in neurons.
Collapse
|
26
|
Hong SK, Kim KH, Song EJ, Kim EE. Structural Basis for the Interaction between the IUS-SPRY Domain of RanBPM and DDX-4 in Germ Cell Development. J Mol Biol 2016; 428:4330-4344. [PMID: 27622290 DOI: 10.1016/j.jmb.2016.09.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/30/2016] [Accepted: 09/02/2016] [Indexed: 02/08/2023]
Abstract
RanBPM and RanBP10 are non-canonical members of the Ran binding protein family that lack the Ran binding domain and do not associate with Ran GTPase in vivo. Rather, they have been shown to be scaffolding proteins that are important for a variety of cellular processes, and both of these proteins contain a SPRY domain, which has been implicated in mediating protein-protein interactions with a variety of targets including the DEAD-box containing ATP-dependent RNA helicase (DDX-4). In this study, we have determined the crystal structures of the SPIa and the ryanodine receptor domain and of approximately 70 upstream residues (immediate upstream to SPRY motif) of both RanBPM and RanBP10. They are almost identical, composed of a β-sandwich fold with a set of two helices on each side located at the edge of the sheets. A unique shallow binding surface is formed by highly conserved loops on the surface of the β-sheet with two aspartates on one end, a positive patch on the opposite end, and a tryptophan lining at the bottom of the surface. The 20-mer peptide (residues 228-247) of human DDX-4, an ATP-dependent RNA helicase known to regulate germ cell development, binds to this surface with a KD of ~13μM. The crystal structure of the peptide complex and the mutagenesis studies elucidate how RanBPM can recognize its interaction partners to function in gametogenesis.
Collapse
Affiliation(s)
- Seung Kon Hong
- Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-gu Hwarang-ro 14-gil 5, Seoul 02792, Republic of Korea
| | - Kook-Han Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-gu Hwarang-ro 14-gil 5, Seoul 02792, Republic of Korea
| | - Eun Joo Song
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seongbuk-gu Hwarang-ro 14-gil 5, Seoul 02792, Republic of Korea
| | - Eunice EunKyeong Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-gu Hwarang-ro 14-gil 5, Seoul 02792, Republic of Korea.
| |
Collapse
|
27
|
Schulte C, Rodighiero S, Cappelluti MA, Puricelli L, Maffioli E, Borghi F, Negri A, Sogne E, Galluzzi M, Piazzoni C, Tamplenizza M, Podestà A, Tedeschi G, Lenardi C, Milani P. Conversion of nanoscale topographical information of cluster-assembled zirconia surfaces into mechanotransductive events promotes neuronal differentiation. J Nanobiotechnology 2016; 14:18. [PMID: 26955876 PMCID: PMC4784317 DOI: 10.1186/s12951-016-0171-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 02/25/2016] [Indexed: 02/03/2023] Open
Abstract
Background Thanks to mechanotransductive components cells are competent to perceive nanoscale topographical features of their environment and to convert the immanent information into corresponding physiological responses. Due to its complex configuration, unraveling the role of the extracellular matrix is particularly challenging. Cell substrates with simplified topographical cues, fabricated by top-down micro- and nanofabrication approaches, have been useful in order to identify basic principles. However, the underlying molecular mechanisms of this conversion remain only partially understood. Results Here we present the results of a broad, systematic and quantitative approach aimed at understanding how the surface nanoscale information is converted into cell response providing a profound causal link between mechanotransductive events, proceeding from the cell/nanostructure interface to the nucleus. We produced nanostructured ZrO2 substrates with disordered yet controlled topographic features by the bottom-up technique supersonic cluster beam deposition, i.e. the assembling of zirconia nanoparticles from the gas phase on a flat substrate through a supersonic expansion. We used PC12 cells, a well-established model in the context of neuronal differentiation. We found that the cell/nanotopography interaction enforces a nanoscopic architecture of the adhesion regions that affects the focal adhesion dynamics and the cytoskeletal organization, which thereby modulates the general biomechanical properties by decreasing the rigidity of the cell. The mechanotransduction impacts furthermore on transcription factors relevant for neuronal differentiation (e.g. CREB), and eventually the protein expression profile. Detailed proteomic data validated the observed differentiation. In particular, the abundance of proteins that are involved in adhesome and/or cytoskeletal organization is striking, and their up- or downregulation is in line with their demonstrated functions in neuronal differentiation processes. Conclusion Our work provides a deep insight into the molecular mechanotransductive mechanisms that realize the conversion of the nanoscale topographical information of SCBD-fabricated surfaces into cellular responses, in this case neuronal differentiation. The results lay a profound cell biological foundation indicating the strong potential of these surfaces in promoting neuronal differentiation events which could be exploited for the development of prospective research and/or biomedical applications. These applications could be e.g. tools to study mechanotransductive processes, improved neural interfaces and circuits, or cell culture devices supporting neurogenic processes. Electronic supplementary material The online version of this article (doi:10.1186/s12951-016-0171-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carsten Schulte
- CIMAINA, Dipartimento di Fisica, Università degli Studi di Milano, via Celoria 16, Milan, 20133, Italy.
| | | | - Martino Alfredo Cappelluti
- SEMM European School of Molecular Medicine, Via Adamello 16, Milan, 20139, Italy. .,Fondazione Filarete, via le Ortles 22/4, Milan, 20139, Italy.
| | - Luca Puricelli
- CIMAINA, Dipartimento di Fisica, Università degli Studi di Milano, via Celoria 16, Milan, 20133, Italy.
| | - Elisa Maffioli
- DIVET, Università degli Studi di Milano, via Celoria 10, Milan, 20133, Italy. .,Fondazione Filarete, via le Ortles 22/4, Milan, 20139, Italy.
| | - Francesca Borghi
- CIMAINA, Dipartimento di Fisica, Università degli Studi di Milano, via Celoria 16, Milan, 20133, Italy.
| | - Armando Negri
- DIVET, Università degli Studi di Milano, via Celoria 10, Milan, 20133, Italy. .,Fondazione Filarete, via le Ortles 22/4, Milan, 20139, Italy.
| | - Elisa Sogne
- SEMM European School of Molecular Medicine, Via Adamello 16, Milan, 20139, Italy. .,Fondazione Filarete, via le Ortles 22/4, Milan, 20139, Italy.
| | - Massimiliano Galluzzi
- CIMAINA, Dipartimento di Fisica, Università degli Studi di Milano, via Celoria 16, Milan, 20133, Italy.
| | - Claudio Piazzoni
- CIMAINA, Dipartimento di Fisica, Università degli Studi di Milano, via Celoria 16, Milan, 20133, Italy.
| | | | - Alessandro Podestà
- CIMAINA, Dipartimento di Fisica, Università degli Studi di Milano, via Celoria 16, Milan, 20133, Italy.
| | - Gabriella Tedeschi
- DIVET, Università degli Studi di Milano, via Celoria 10, Milan, 20133, Italy. .,Fondazione Filarete, via le Ortles 22/4, Milan, 20139, Italy.
| | - Cristina Lenardi
- CIMAINA, Dipartimento di Fisica, Università degli Studi di Milano, via Celoria 16, Milan, 20133, Italy.
| | - Paolo Milani
- CIMAINA, Dipartimento di Fisica, Università degli Studi di Milano, via Celoria 16, Milan, 20133, Italy.
| |
Collapse
|
28
|
Cellular Transcriptional Coactivator RanBP10 and Herpes Simplex Virus 1 ICP0 Interact and Synergistically Promote Viral Gene Expression and Replication. J Virol 2016; 90:3173-86. [PMID: 26739050 DOI: 10.1128/jvi.03043-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 12/30/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED To investigate the molecular mechanism(s) by which herpes simplex virus 1 (HSV-1) regulatory protein ICP0 promotes viral gene expression and replication, we screened cells overexpressing ICP0 for ICP0-binding host cell proteins. Tandem affinity purification of transiently expressed ICP0 coupled with mass spectrometry-based proteomics technology and subsequent analyses showed that ICP0 interacted with cell protein RanBP10, a known transcriptional coactivator, in HSV-1-infected cells. Knockdown of RanBP10 in infected HEp-2 cells resulted in a phenotype similar to that observed with the ICP0-null mutation, including reduction in viral replication and in the accumulation of viral immediate early (ICP27), early (ICP8), and late (VP16) mRNAs and proteins. In addition, RanBP10 knockdown or the ICP0-null mutation increased the level of histone H3 association with the promoters of these viral genes, which is known to repress transcription. These effects observed in wild-type HSV-1-infected HEp-2 RanBP10 knockdown cells or those observed in ICP0-null mutant virus-infected control HEp-2 cells were remarkably increased in ICP0-null mutant virus-infected HEp-2 RanBP10 knockdown cells. Our results suggested that ICP0 and RanBP10 redundantly and synergistically promoted viral gene expression by regulating chromatin remodeling of the HSV-1 genome for efficient viral replication. IMPORTANCE Upon entry of herpesviruses into a cell, viral gene expression is restricted by heterochromatinization of the viral genome. Therefore, HSV-1 has evolved multiple mechanisms to counteract this epigenetic silencing for efficient viral gene expression and replication. HSV-1 ICP0 is one of the viral proteins involved in counteracting epigenetic silencing. Here, we identified RanBP10 as a novel cellular ICP0-binding protein and showed that RanBP10 and ICP0 appeared to act synergistically to promote viral gene expression and replication by modulating viral chromatin remodeling. Our results provide insight into the mechanisms by which HSV-1 regulates viral chromatin remodeling for efficient viral gene expression and replication.
Collapse
|
29
|
Twiss JL, Kalinski AL, Sachdeva R, Houle JD. Intra-axonal protein synthesis - a new target for neural repair? Neural Regen Res 2016; 11:1365-1367. [PMID: 27857722 PMCID: PMC5090821 DOI: 10.4103/1673-5374.191193] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Although initially argued to be a feature of immature neurons with incomplete polarization, there is clear evidence that neurons in the peripheral nervous system retain the capacity for intra-axonal protein synthesis well into adulthood. This localized protein synthesis has been shown to contribute to injury signaling and axon regeneration in peripheral nerves. Recent works point to potential for protein synthesis in axons of the vertebrate central nervous system. mRNAs and protein synthesis machinery have now been documented in lamprey, mouse, and rat spinal cord axons. Intra-axonal protein synthesis appears to be activated in adult vertebrate spinal cord axons when they are regeneration-competent. Rat spinal cord axons regenerating into a peripheral nerve graft contain mRNAs and markers of activated translational machinery. Indeed, levels of some growth-associated mRNAs in these spinal cord axons are comparable to the regenerating sciatic nerve. Markers of active translation tend to decrease when these axons stop growing, but can be reactivated by a second axotomy. These emerging observations raise the possibility that mRNA transport into and translation within axons could be targeted to facilitate regeneration in both the peripheral and central nervous systems.
Collapse
Affiliation(s)
- Jeffery L Twiss
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
| | - Ashley L Kalinski
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
| | - Rahul Sachdeva
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - John D Houle
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
30
|
Simon S, Hilbi H. Subversion of Cell-Autonomous Immunity and Cell Migration by Legionella pneumophila Effectors. Front Immunol 2015; 6:447. [PMID: 26441958 PMCID: PMC4568765 DOI: 10.3389/fimmu.2015.00447] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/17/2015] [Indexed: 12/14/2022] Open
Abstract
Bacteria trigger host defense and inflammatory processes, such as cytokine production, pyroptosis, and the chemotactic migration of immune cells toward the source of infection. However, a number of pathogens interfere with these immune functions by producing specific so-called “effector” proteins, which are delivered to host cells via dedicated secretion systems. Air-borne Legionella pneumophila bacteria trigger an acute and potential fatal inflammation in the lung termed Legionnaires’ disease. The opportunistic pathogen L. pneumophila is a natural parasite of free-living amoebae, but also replicates in alveolar macrophages and accidentally infects humans. The bacteria employ the intracellular multiplication/defective for organelle trafficking (Icm/Dot) type IV secretion system and as many as 300 different effector proteins to govern host–cell interactions and establish in phagocytes an intracellular replication niche, the Legionella-containing vacuole. Some Icm/Dot-translocated effector proteins target cell-autonomous immunity or cell migration, i.e., they interfere with (i) endocytic, secretory, or retrograde vesicle trafficking pathways, (ii) organelle or cell motility, (iii) the inflammasome and programed cell death, or (iv) the transcription factor NF-κB. Here, we review recent mechanistic insights into the subversion of cellular immune functions by L. pneumophila.
Collapse
Affiliation(s)
- Sylvia Simon
- Institute of Medical Microbiology, University of Zürich , Zürich , Switzerland
| | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zürich , Zürich , Switzerland ; Max von Pettenkofer Institute, Ludwig-Maximilians University , Munich , Germany
| |
Collapse
|
31
|
Dörr A, Pierre S, Zhang DD, Henke M, Holland S, Scholich K. MYCBP2 Is a Guanosine Exchange Factor for Ran Protein and Determines Its Localization in Neurons of Dorsal Root Ganglia. J Biol Chem 2015; 290:25620-35. [PMID: 26304119 DOI: 10.1074/jbc.m115.646901] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Indexed: 11/06/2022] Open
Abstract
The small GTPase Ran coordinates retrograde axonal transport in neurons, spindle assembly during mitosis, and the nucleo-cytoplasmic transport of mRNA. Its localization is tightly regulated by the GTPase-activating protein RanGAP1 and the nuclear guanosine exchange factor (GEF) RCC1. We show that loss of the neuronal E3 ubiquitin ligase MYCBP2 caused the up-regulation of Ran and RanGAP1 in dorsal root ganglia (DRG) under basal conditions and during inflammatory hyperalgesia. SUMOylated RanGAP1 physically interacted with MYCBP2 and inhibited its E3 ubiquitin ligase activity. Stimulation of neurons induced a RanGAP1-dependent translocation of MYCBP2 to the nucleus. In the nucleus of DRG neurons MYCBP2 co-localized with Ran and facilitated through its RCC1-like domain the GDP/GTP exchange of Ran. In accordance with the necessity of a GEF to promote GTP-binding and nuclear export of Ran, the nuclear localization of Ran was strongly increased in MYCBP2-deficient DRGs. The finding that other GEFs for Ran besides RCC1 exist gives new insights in the complexity of the regulation of the Ran signaling pathway.
Collapse
Affiliation(s)
- Angela Dörr
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, Frankfurt 60590, Germany
| | - Sandra Pierre
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, Frankfurt 60590, Germany
| | - Dong D Zhang
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, Frankfurt 60590, Germany
| | - Marina Henke
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, Frankfurt 60590, Germany
| | - Sabrina Holland
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, Frankfurt 60590, Germany
| | - Klaus Scholich
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, Frankfurt 60590, Germany
| |
Collapse
|
32
|
Abstract
Neurons are extremely polarized cells. Axon lengths often exceed the dimension of the neuronal cell body by several orders of magnitude. These extreme axonal lengths imply that neurons have mastered efficient mechanisms for long distance signaling between soma and synaptic terminal. These elaborate mechanisms are required for neuronal development and maintenance of the nervous system. Neurons can fine-tune long distance signaling through calcium wave propagation and bidirectional transport of proteins, vesicles, and mRNAs along microtubules. The signal transmission over extreme lengths also ensures that information about axon injury is communicated to the soma and allows for repair mechanisms to be engaged. This review focuses on the different mechanisms employed by neurons to signal over long axonal distances and how signals are interpreted in the soma, with an emphasis on proteomic studies. We also discuss how proteomic approaches could help further deciphering the signaling mechanisms operating over long distance in axons.
Collapse
Affiliation(s)
- Atsushi Saito
- From the ‡Department of Anatomy and Neurobiology, Washington University in St Louis, School of Medicine, St Louis, 63110, Missouri
| | - Valeria Cavalli
- From the ‡Department of Anatomy and Neurobiology, Washington University in St Louis, School of Medicine, St Louis, 63110, Missouri.
| |
Collapse
|
33
|
Hilbi H, Rothmeier E, Hoffmann C, Harrison CF. Beyond Rab GTPases Legionella activates the small GTPase Ran to promote microtubule polymerization, pathogen vacuole motility, and infection. Small GTPases 2015; 5:1-6. [PMID: 25496424 DOI: 10.4161/21541248.2014.972859] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Legionella spp. are amoebae-resistant environmental bacteria that replicate in free-living protozoa in a distinct compartment, the Legionella-containing vacuole (LCV). Upon transmission of Legionella pneumophila to the lung, the pathogens employ an evolutionarily conserved mechanism to grow in LCVs within alveolar macrophages, thus triggering a severe pneumonia termed Legionnaires' disease. LCV formation is a complex and robust process, which requires the bacterial Icm/Dot type IV secretion system and involves the amazing number of 300 different translocated effector proteins. LCVs interact with the host cell's endosomal and secretory vesicle trafficking pathway. Accordingly, in a proteomics approach as many as 12 small Rab GTPases implicated in endosomal and secretory vesicle trafficking were identified and validated as LCV components. Moreover, the small GTPase Ran and its effector protein RanBP1 have been found to decorate the pathogen vacuole. Ran regulates nucleo-cytoplasmic transport, spindle assembly, and cytokinesis, as well as the organization of non-centrosomal microtubules. In L. pneumophila-infected amoebae or macrophages, Ran and RanBP1 localize to LCVs, and the small GTPase is activated by the Icm/Dot substrate LegG1. Ran activation by LegG1 leads to microtubule stabilization and promotes intracellular pathogen vacuole motility and bacterial growth, as well as chemotaxis and migration of Legionella-infected cells.
Collapse
Affiliation(s)
- Hubert Hilbi
- a Max von Pettenkofer Institute; Department of Medicine ; Ludwig-Maximilians University Munich ; Munich , Germany
| | | | | | | |
Collapse
|
34
|
Joy T, Hirono K, Doe CQ. The RanGEF Bj1 promotes prospero nuclear export and neuroblast self-renewal. Dev Neurobiol 2014; 75:485-93. [PMID: 25312250 PMCID: PMC4397115 DOI: 10.1002/dneu.22237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 10/03/2014] [Accepted: 10/09/2014] [Indexed: 11/08/2022]
Abstract
Drosophila larval neuroblasts are a model system for studying stem cell self-renewal and differentiation. Here, we report a novel role for the Drosophila gene Bj1 in promoting larval neuroblast self-renewal. Bj1 is the guanine-nucleotide exchange factor for Ran GTPase, which regulates nuclear import/export. Bj1 transcripts are highly enriched in larval brain neuroblasts (in both central brain and optic lobe), while Bj1 protein is detected in both neuroblasts and their neuronal progeny. Loss of Bj1 using both mutants or RNAi causes a progressive loss of larval neuroblasts, showing that Bj1 is required to maintain neuroblast numbers. Loss of Bj1 does not result in neuroblast apoptosis, but rather leads to abnormal nuclear accumulation of the differentiation factor Prospero, and premature neuroblast differentiation. We conclude that the Bj1 RanGEF promotes Prospero nuclear export and neuroblast self-renewal.
Collapse
Affiliation(s)
- Tasha Joy
- Institute of Molecular Biology, Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, Oregon, 97403
| | | | | |
Collapse
|
35
|
Abstract
The Ras superfamily of small GTPases is composed of more than 150 members, which share a conserved structure and biochemical properties, acting as binary molecular switches turned on by binding GTP and off by hydrolyzing GTP to GDP. However, despite considerable structural and biochemical similarities, these proteins play multiple and divergent roles, being versatile and key regulators of virtually all fundamental cellular processes. Conversely, their dysfunction plays a crucial role in the pathogenesis of serious human diseases, including cancer and developmental syndromes. Fuelled by the original identification in 1982 of mutationally activated and transforming human Ras genes in human cancer cell lines, a variety of powerful experimental techniques have been intensively focused on discovering and studying structure, biochemistry, and biology of Ras and Ras-related small GTPases, leading to fundamental research breakthroughs into identification and structural and functional characterization of a huge number of Ras superfamily members, as well as of their multiple regulators and effectors. In this review we provide a general overview of the major milestones that eventually allowed to unlock the secret treasure chest of this large and important superfamily of proteins.
Collapse
|
36
|
Brasseur A, Bayat S, Chua XL, Zhang Y, Zhou Q, Low BC, He CY. The bi-lobe-associated LRRP1 regulates Ran activity in Trypanosoma brucei. J Cell Sci 2014; 127:4846-56. [PMID: 25217630 DOI: 10.1242/jcs.148015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Cilia and flagella are conserved eukaryotic organelles important for motility and sensory. The RanGTPase, best known for nucleocytoplasmic transport functions, may also play a role in protein trafficking into the specialized flagellar/ciliary compartments, although the regulatory mechanisms controlling Ran activity at the flagellum remain unclear. The unicellular parasite Trypanosoma brucei contains a single flagellum necessary for cell movement, division and morphogenesis. Correct flagellum functions require flagellar attachment to the cell body, which is mediated by a specialized flagellum attachment zone (FAZ) complex that is assembled together with the flagellum during the cell cycle. We have previously identified the leucine-rich-repeat protein 1 LRRP1 on a bi-lobe structure at the proximal base of flagellum and FAZ. LRRP1 is essential for bi-lobe and FAZ biogenesis, consequently affecting flagellum-driven cell motility and division. Here, we show that LRRP1 forms a complex with Ran and a Ran-binding protein, and regulates Ran-GTP hydrolysis in T. brucei. In addition to mitotic inhibition, depletion of Ran inhibits FAZ assembly in T. brucei, supporting the presence of a conserved mechanism that involves Ran in the regulation of flagellum functions in an early divergent eukaryote.
Collapse
Affiliation(s)
- Anaïs Brasseur
- Department of Biological Sciences, Centre for BioImaging Sciences, National University of Singapore, 14 Science Drive 4, S1A #06-04, Singapore 117543, Singapore
| | - Shima Bayat
- Department of Biological Sciences, Centre for BioImaging Sciences, National University of Singapore, 14 Science Drive 4, S1A #06-04, Singapore 117543, Singapore
| | - Xiu Ling Chua
- Department of Biological Sciences, Centre for BioImaging Sciences, National University of Singapore, 14 Science Drive 4, S1A #06-04, Singapore 117543, Singapore
| | - Yu Zhang
- Department of Biological Sciences, Centre for BioImaging Sciences, National University of Singapore, 14 Science Drive 4, S1A #06-04, Singapore 117543, Singapore
| | | | - Boon Chuan Low
- Singapore Mechanobiology Institute, National University of Singapore, T-Lab, #10-01, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Cynthia Y He
- Department of Biological Sciences, Centre for BioImaging Sciences, National University of Singapore, 14 Science Drive 4, S1A #06-04, Singapore 117543, Singapore
| |
Collapse
|
37
|
Liu P, Qi M, Wang Y, Chang M, Liu C, Sun M, Yang W, Ren H. Arabidopsis RAN1 mediates seed development through its parental ratio by affecting the onset of endosperm cellularization. MOLECULAR PLANT 2014; 7:1316-1328. [PMID: 24719465 DOI: 10.1093/mp/ssu041] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Although previous studies have demonstrated that endosperm development is influenced by its parental genome constitution, the genetic basis and molecular mechanisms that control parent-of-origin effects require further elucidation. Here we show that the Ras-related nuclear protein 1 (RAN1) regulates endosperm development in Arabidopsis thaliana. Reciprocal crosses between wild-type (WT) and transgenic lines misexpressing RAN1 (msRAN1) gave rise to small F1 seeds when RAN1 down-regulated/up-regulated individuals were used as a male/female parent; in contrast, F1 seeds were aborted when RAN1 down-regulated/up-regulated plants were used as a female/male parent, suggesting that seed development is affected by the parental genome ratio of RAN1. Whereas RAN1 expression in wild-type plants is reduced before the onset of endosperm cellularization, F1 seeds from reciprocal crosses between WT and msRAN1 showed abnormal endosperm cellularization and ectopic expression of RAN1. The expression of MINISEED3 (MINI3)-a gene that also controls endosperm cellularization-was also affected in these reciprocal crosses, and the misregulation of MINI3 activity rescued F1 seeds when msRAN1 plants were used in reciprocal crosses. Taken together, our results suggest that the parental ratio of RAN1 regulates the onset of endosperm cellularization through its genetic interaction with MINI3.
Collapse
Affiliation(s)
- Peiwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Ming Qi
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yuqian Wang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Mingqin Chang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Chang Liu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Mengxiang Sun
- Department of Cell and Development Biology, College of Life Sciences, State Key Laboratory of Hybrid Rice, Wuhan University, Wuhan 430072, China
| | - Weicai Yang
- Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Haiyun Ren
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
38
|
Igarashi M. Proteomic identification of the molecular basis of mammalian CNS growth cones. Neurosci Res 2014; 88:1-15. [PMID: 25066522 DOI: 10.1016/j.neures.2014.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/13/2014] [Accepted: 07/02/2014] [Indexed: 11/28/2022]
Abstract
The growth cone, which is a unique structure with high motility that forms at the tips of extending axons and dendrites, is crucial to neuronal network formation. Axonal growth of the mammalian CNS is most likely achieved by the complicated coordination of cytoskeletal rearrangement and vesicular trafficking via many proteins. Before recent advances, no methods to identify numerous proteins existed; however, proteomics revolutionarily resolved such problems. In this review, I summarize the profiles of the mammalian growth cone proteins revealed by proteomics as the molecular basis of the growth cone functions, with molecular mapping. These results should be used as a basis for understanding the mechanisms of the complex mammalian CNS developmental process.
Collapse
Affiliation(s)
- Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Trans-disciplinary Program, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
39
|
Hilbi H, Rothmeier E, Hoffmann C, Harrison CF. Beyond Rab GTPases: Legionella activates the small GTPase Ran to promote microtubule polymerization, pathogen vacuole motility, and infection. Small GTPases 2014. [DOI: 10.4161/sgtp.28651] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
40
|
Kim YI, Bandyopadhyay J, Cho I, Lee J, Park DH, Cho JH. Nucleolar GTPase NOG-1 regulates development, fat storage, and longevity through insulin/IGF signaling in C. elegans. Mol Cells 2014; 37:51-7. [PMID: 24552710 PMCID: PMC3907010 DOI: 10.14348/molcells.2014.2251] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 11/28/2013] [Accepted: 11/29/2013] [Indexed: 12/11/2022] Open
Abstract
NOG1 is a nucleolar GTPase that is critical for 60S ribosome biogenesis. Recently, NOG1 was identified as one of the downstream regulators of target of rapamycin (TOR) in yeast. It is reported that TOR is involved in regulating lifespan and fat storage in Caenorhabditis elegans. Here, we show that the nog1 ortholog (T07A9.9: nog-1) in C. elegans regulates growth, development, lifespan, and fat metabolism. A green fluorescence protein (GFP) promoter assay revealed ubiquitous expression of C. elegans nog-1 from the early embryonic to the adult stage. Furthermore, the GFP-tagged NOG-1 protein is localized to the nucleus, whereas the aberrant NOG-1 protein is concentrated in the nucleolus. Functional studies of NOG-1 in C. elegans further revealed that nog-1 knockdown resulted in smaller broodsize, slower growth, increased life span, and more fat storage. Moreover, nog-1 over-expression resulted in decreased life span. Taken together, our data suggest that nog-1 in C. elegans may be an important player in regulating life span and fat storage via the insulin/IGF pathway.
Collapse
Affiliation(s)
- Young-Il Kim
- Biomedical Research Center, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Jaya Bandyopadhyay
- Department of Biotechnology, West Bengal University of Technology, Salt Lake City, Kolkata 700-064,
India
| | - Injeong Cho
- Department of Biology Education, College of Education, Chosun University, Gwangju 501-759,
Korea
| | - Juyeon Lee
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712,
Korea
| | - Dae Ho Park
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712,
Korea
| | - Jeong Hoon Cho
- Department of Biology Education, College of Education, Chosun University, Gwangju 501-759,
Korea
| |
Collapse
|
41
|
Abstract
The extensive lengths of neuronal processes necessitate efficient mechanisms for communication with the cell body. Neuronal regeneration after nerve injury requires new transcription; thus, long-distance retrograde signalling from axonal lesion sites to the soma and nucleus is required. In recent years, considerable progress has been made in elucidating the mechanistic basis of this system. This has included the discovery of a priming role for early calcium waves; confirmation of central roles for mitogen-activated protein kinase signalling effectors, the importin family of nucleocytoplasmic transport factors and molecular motors such as dynein; and demonstration of the importance of local translation as a key regulatory mechanism. These recent findings provide a coherent mechanistic framework for axon-soma communication in the injured nerve and shed light on the integration of cytoplasmic and nuclear transport in all eukaryotic cells.
Collapse
Affiliation(s)
- Ida Rishal
- Department of Biological Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Mike Fainzilber
- Department of Biological Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
42
|
Citalán-Madrid AF, García-Ponce A, Vargas-Robles H, Betanzos A, Schnoor M. Small GTPases of the Ras superfamily regulate intestinal epithelial homeostasis and barrier function via common and unique mechanisms. Tissue Barriers 2013; 1:e26938. [PMID: 24868497 PMCID: PMC3942330 DOI: 10.4161/tisb.26938] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 10/21/2013] [Accepted: 10/24/2013] [Indexed: 12/11/2022] Open
Abstract
The intestinal epithelium forms a stable barrier protecting underlying tissues from pathogens in the gut lumen. This is achieved by specialized integral membrane structures such as tight and adherens junctions that connect neighboring cells and provide stabilizing links to the cytoskeleton. Junctions are constantly remodeled to respond to extracellular stimuli. Assembly and disassembly of junctions is regulated by interplay of actin remodeling, endocytotic recycling of junctional proteins, and various signaling pathways. Accumulating evidence implicate small G proteins of the Ras superfamily as important signaling molecules for the regulation of epithelial junctions. They function as molecular switches circling between an inactive GDP-bound and an active GTP-bound state. Once activated, they bind different effector molecules to control cellular processes required for correct junction assembly, maintenance and remodelling. Here, we review recent advances in understanding how GTPases of the Rho, Ras, Rab and Arf families contribute to intestinal epithelial homeostasis.
Collapse
Affiliation(s)
- Alí Francisco Citalán-Madrid
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Alexander García-Ponce
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Hilda Vargas-Robles
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Abigail Betanzos
- Department of Infectomics and Molecular Pathogenesis; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| |
Collapse
|
43
|
Vyas P, Singh A, Murawala P, Joseph J. Nup358 interacts with Dishevelled and aPKC to regulate neuronal polarity. Biol Open 2013; 2:1270-8. [PMID: 24244865 PMCID: PMC3828775 DOI: 10.1242/bio.20135363] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 10/01/2013] [Indexed: 11/25/2022] Open
Abstract
Par polarity complex, consisting of Par3, Par6, and aPKC, plays a conserved role in the establishment and maintenance of polarization in diverse cellular contexts. Recent reports suggest that Dishevelled (Dvl), a cytoplasmic mediator of Wnt signalling, interacts with atypical protein kinase C and regulates its activity during neuronal differentiation and directed cell migration. Here we show that Nup358 (also called RanBP2), a nucleoporin previously implicated in polarity during directed cell migration, interacts with Dishevelled and aPKC through its N-terminal region (BPN) and regulates axon–dendrite differentiation of cultured hippocampal neurons. Depletion of endogenous Nup358 leads to generation of multiple axons, whereas overexpression of BPN abrogates the process of axon formation. Moreover, siRNA-mediated knockdown of Dvl or inhibition of aPKC by a pseudosubstrate inhibitor significantly reverses the multiple axon phenotype produced by Nup358 depletion. Collectively, these data suggest that Nup358 plays an important role in regulating neuronal polarization upstream to Dvl and aPKC.
Collapse
Affiliation(s)
- Pankhuri Vyas
- National Centre for Cell Science , Ganeshkhind, Pune 411007, Maharashtra , India
| | | | | | | |
Collapse
|
44
|
Loirand G, Sauzeau V, Pacaud P. Small G Proteins in the Cardiovascular System: Physiological and Pathological Aspects. Physiol Rev 2013; 93:1659-720. [DOI: 10.1152/physrev.00021.2012] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Small G proteins exist in eukaryotes from yeast to human and constitute the Ras superfamily comprising more than 100 members. This superfamily is structurally classified into five families: the Ras, Rho, Rab, Arf, and Ran families that control a wide variety of cell and biological functions through highly coordinated regulation processes. Increasing evidence has accumulated to identify small G proteins and their regulators as key players of the cardiovascular physiology that control a large panel of cardiac (heart rhythm, contraction, hypertrophy) and vascular functions (angiogenesis, vascular permeability, vasoconstriction). Indeed, basal Ras protein activity is required for homeostatic functions in physiological conditions, but sustained overactivation of Ras proteins or spatiotemporal dysregulation of Ras signaling pathways has pathological consequences in the cardiovascular system. The primary object of this review is to provide a comprehensive overview of the current progress in our understanding of the role of small G proteins and their regulators in cardiovascular physiology and pathologies.
Collapse
Affiliation(s)
- Gervaise Loirand
- INSERM, UMR S1087; University of Nantes; and CHU Nantes, l'Institut du Thorax, Nantes, France
| | - Vincent Sauzeau
- INSERM, UMR S1087; University of Nantes; and CHU Nantes, l'Institut du Thorax, Nantes, France
| | - Pierre Pacaud
- INSERM, UMR S1087; University of Nantes; and CHU Nantes, l'Institut du Thorax, Nantes, France
| |
Collapse
|
45
|
Identification of a PTC-containing DlRan transcript and its differential expression during somatic embryogenesis in Dimocarpus longan. Gene 2013; 529:37-44. [DOI: 10.1016/j.gene.2013.07.091] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/19/2013] [Accepted: 07/25/2013] [Indexed: 11/17/2022]
|
46
|
Rothmeier E, Pfaffinger G, Hoffmann C, Harrison CF, Grabmayr H, Repnik U, Hannemann M, Wölke S, Bausch A, Griffiths G, Müller-Taubenberger A, Itzen A, Hilbi H. Activation of Ran GTPase by a Legionella effector promotes microtubule polymerization, pathogen vacuole motility and infection. PLoS Pathog 2013; 9:e1003598. [PMID: 24068924 PMCID: PMC3777869 DOI: 10.1371/journal.ppat.1003598] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 07/22/2013] [Indexed: 11/19/2022] Open
Abstract
The causative agent of Legionnaires' disease, Legionella pneumophila, uses the Icm/Dot type IV secretion system (T4SS) to form in phagocytes a distinct "Legionella-containing vacuole" (LCV), which intercepts endosomal and secretory vesicle trafficking. Proteomics revealed the presence of the small GTPase Ran and its effector RanBP1 on purified LCVs. Here we validate that Ran and RanBP1 localize to LCVs and promote intracellular growth of L. pneumophila. Moreover, the L. pneumophila protein LegG1, which contains putative RCC1 Ran guanine nucleotide exchange factor (GEF) domains, accumulates on LCVs in an Icm/Dot-dependent manner. L. pneumophila wild-type bacteria, but not strains lacking LegG1 or a functional Icm/Dot T4SS, activate Ran on LCVs, while purified LegG1 produces active Ran(GTP) in cell lysates. L. pneumophila lacking legG1 is compromised for intracellular growth in macrophages and amoebae, yet is as cytotoxic as the wild-type strain. A downstream effect of LegG1 is to stabilize microtubules, as revealed by conventional and stimulated emission depletion (STED) fluorescence microscopy, subcellular fractionation and Western blot, or by microbial microinjection through the T3SS of a Yersinia strain lacking endogenous effectors. Real-time fluorescence imaging indicates that LCVs harboring wild-type L. pneumophila rapidly move along microtubules, while LCVs harboring ΔlegG1 mutant bacteria are stalled. Together, our results demonstrate that Ran activation and RanBP1 promote LCV formation, and the Icm/Dot substrate LegG1 functions as a bacterial Ran activator, which localizes to LCVs and promotes microtubule stabilization, LCV motility as well as intracellular replication of L. pneumophila.
Collapse
Affiliation(s)
- Eva Rothmeier
- Max von Pettenkofer-Institute, Department of Medicine, Ludwig-Maximilians Universität München, München, Germany
| | - Gudrun Pfaffinger
- Max von Pettenkofer-Institute, Department of Medicine, Ludwig-Maximilians Universität München, München, Germany
| | - Christine Hoffmann
- Max von Pettenkofer-Institute, Department of Medicine, Ludwig-Maximilians Universität München, München, Germany
| | - Christopher F. Harrison
- Max von Pettenkofer-Institute, Department of Medicine, Ludwig-Maximilians Universität München, München, Germany
| | - Heinrich Grabmayr
- Institute of Molecular and Cellular Biophysics, Department of Physics, Technische Universität München, Garching, Germany
| | - Urska Repnik
- Department of Molecular Biosciences, University of Oslo, Oslo, Norway
| | - Mandy Hannemann
- Center for Integrated Protein Science Munich, Department of Chemistry, Technische Universität München, Garching, Germany
| | - Stefan Wölke
- Max von Pettenkofer-Institute, Department of Medicine, Ludwig-Maximilians Universität München, München, Germany
| | - Andreas Bausch
- Institute of Molecular and Cellular Biophysics, Department of Physics, Technische Universität München, Garching, Germany
| | - Gareth Griffiths
- Department of Molecular Biosciences, University of Oslo, Oslo, Norway
| | - Annette Müller-Taubenberger
- Institute for Anatomy and Cell Biology, Department of Medicine, Ludwig-Maximilians Universität München, München, Germany
| | - Aymelt Itzen
- Center for Integrated Protein Science Munich, Department of Chemistry, Technische Universität München, Garching, Germany
| | - Hubert Hilbi
- Max von Pettenkofer-Institute, Department of Medicine, Ludwig-Maximilians Universität München, München, Germany
- * E-mail:
| |
Collapse
|
47
|
Targeting the Dbl and dock-family RhoGEFs: a yeast-based assay to identify cell-active inhibitors of Rho-controlled pathways. Enzymes 2013; 33 Pt A:169-91. [PMID: 25033805 DOI: 10.1016/b978-0-12-416749-0.00008-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The Ras-like superfamily of low molecular weight GTPases is made of five major families (Arf/Sar, Rab, Ran, Ras, and Rho), highly conserved across evolution. This is in keeping with their roles in basic cellular functions (endo/exocytosis, vesicular trafficking, nucleocytoplasmic trafficking, cell signaling, proliferation and apoptosis, gene regulation, F-actin dynamics), whose alterations are associated with various types of diseases, in particular cancer, neurodegenerative, cardiovascular, and infectious diseases. For these reasons, Ras-like pathways are of great potential in therapeutics and identifying inhibitors that decrease signaling activity is under intense research. Along this line, guanine exchange factors (GEFs) represent attractive targets. GEFs are proteins that promote the active GTP-bound state of GTPases and represent the major entry points whereby extracellular cues are converted into Ras-like signaling. We previously developed the yeast exchange assay (YEA), an experimental setup in the yeast in which activity of a mammalian GEF can be monitored by auxotrophy and color reporter genes. This assay was further engineered for medium-throughput screening of GEF inhibitors, which can readily select for cell-active and specific compounds. We report here on the successful identification of inhibitors against Dbl and CZH/DOCK-family members, GEFs for Rho GTPases, and on the experimental setup to screen for inhibitors of GEFs of the Arf family. We also discuss on inhibitors developed using virtual screening (VS), which target the GEF/GTPase interface with high efficacy and specificity. We propose that using VS and YEA in combination may represent a method of choice for identifying specific and cell-active GEF inhibitors.
Collapse
|
48
|
Liang F, Li L, Wang M, Niu X, Zhan J, He X, Yu C, Jiang M, Lu A. Molecular network and chemical fragment-based characteristics of medicinal herbs with cold and hot properties from Chinese medicine. JOURNAL OF ETHNOPHARMACOLOGY 2013; 148:770-9. [PMID: 23702041 DOI: 10.1016/j.jep.2013.04.055] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 04/15/2013] [Accepted: 04/28/2013] [Indexed: 05/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicines (HMs) is one of the great herbal systems of the world, which play an important role in current health care system in many countries. In the view of tradition Chinese medicine (TCM) theory, Yin-yang and five-elements theory is the central theory, which is used to explain how the world and body work. Under the guidance of such philosophy, TCM considers that HMs have different properties, which are the important factors for prescribing herbal formulae; such prescriptions are based on TCM pattern classification in clinical practice. The cold and hot property are commonly defined for HM property identification; however, the biological activities that are related to the HM property remain a mystery because of a lack of appropriate methods. A bioinformatics approach was applied to identify the distinguishing biological activities of HMs that have these cold and hot properties. MATERIAL AND METHODS Twenty HMs with typical cold and hot properties (10 cold and 10 hot) were selected based on TCM clinical application records and Chinese pharmacopeia. The active target proteins of each HM were searched in the PubChem database and were analyzed in Ingenuity Pathway Analysis (IPA) platform to find out the HM property-related biological activities. In addition, the main compounds of the HMs were fragmented using a fragment-based approach and were analyzed for the purpose of deciphering the properties. RESULTS The main biological networks of HMs with cold and hot properties include cell cycle, cellular growth, proliferation and development, cancer, cytokine signaling, and intracellular and second messenger signaling; 11 specific pathways are presented to be perturbed only by HMs with the hot property, and the 27 specific target protein molecules include PRKACA, PRKCA, PRKCB, PRKCD, PRKCE, PRKCG, PRKD1, TLR4, TLR7, TLR8, TLR9, HTR4, HTR6, HTR7, HTR2A, HTR1B, HTR2B, GNAO1, GNAI1, TNF, IL8, ROCK2, AKT1, MAPK1, RPS6KA1, RPS6KA3 and JAK2, which are involved in the biological network. One specific pathway is detected to be involved in the biological network of HMs with the cold property, the specific molecules are RAN and KPNB1. Cold propertied HMs show intensive toxicity in the heart, liver and kidney compared with hot HMs, which is likely to be correlated with the specific chemical fragments constructions in the HMs with the cold property, such as long chain alkenes, Benzo heterocycle and azotic heterocycle according to the chemical fragment analysis for the HMs. CONCLUSIONS Inflammation and immunity regulation are more related to HMs with the hot property, and cold propertied HMs possess the tendency to impact cell growth, proliferation and development. Integrative bioinformatics analysis and chemical structure analysis are a promising methods for identifying the biological activity of HM properties.
Collapse
Affiliation(s)
- Fei Liang
- Institute of Basic Research In Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing 100700, China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Shin HY, Reich NC. Dynamic trafficking of STAT5 depends on an unconventional nuclear localization signal. J Cell Sci 2013; 126:3333-43. [PMID: 23704351 DOI: 10.1242/jcs.123042] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Signal transducer and activator of transcription 5 (STAT5) is crucial for physiological processes that include hematopoiesis, liver metabolism and mammary gland development. However, aberrant continual activity of STAT5 has been causally linked to human leukemias and solid tumor formation. As a regulated transcription factor, precise cellular localization of STAT5 is essential. Conventional nuclear localization signals consist of short stretches of basic amino acids. In this study, we provide evidence that STAT5 nuclear import is dependent on an unconventional nuclear localization signal that functions within the conformation of an extensive coiled-coil domain. Both in vitro binding and in vivo functional assays reveal that STAT5 nuclear import is mediated by the importin-α3/β1 system independently of STAT5 activation by tyrosine phosphorylation. The integrity of the coiled-coil domain is essential for STAT5 transcriptional induction of the β-casein gene following prolactin stimulation as well as its ability to synergize with the glucocorticoid receptor. The glucocorticoid receptor accumulates in the nucleus in response to prolactin and this nuclear import is dependent on STAT5 nuclear import. STAT5 continually shuttles in and out of the nucleus and live cell imaging demonstrates that STAT5 nuclear export is mediated by both chromosome region maintenance 1 (Crm1)-dependent and Crm1-independent pathways. A Crm1-dependent nuclear export signal was identified within the STAT5 N-terminus. These findings provide insight into the fundamental mechanisms that regulate STAT5 nuclear trafficking and cooperation with the glucocorticoid receptor and provide a basis for clinical intervention of STAT5 function in disease.
Collapse
Affiliation(s)
- Ha Youn Shin
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794, USA
| | | |
Collapse
|
50
|
Abstract
Small GTPases use GDP/GTP alternation to actuate a variety of functional switches that are pivotal for cell dynamics. The GTPase switch is turned on by GEFs, which stimulate dissociation of the tightly bound GDP, and turned off by GAPs, which accelerate the intrinsically sluggish hydrolysis of GTP. For Ras, Rho, and Rab GTPases, this switch incorporates a membrane/cytosol alternation regulated by GDIs and GDI-like proteins. The structures and core mechanisms of representative members of small GTPase regulators from most families have now been elucidated, illuminating their general traits combined with scores of unique features. Recent studies reveal that small GTPase regulators have themselves unexpectedly sophisticated regulatory mechanisms, by which they process cellular signals and build up specific cell responses. These mechanisms include multilayered autoinhibition with stepwise release, feedback loops mediated by the activated GTPase, feed-forward signaling flow between regulators and effectors, and a phosphorylation code for RhoGDIs. The flipside of these highly integrated functions is that they make small GTPase regulators susceptible to biochemical abnormalities that are directly correlated with diseases, notably a striking number of missense mutations in congenital diseases, and susceptible to bacterial mimics of GEFs, GAPs, and GDIs that take command of small GTPases in infections. This review presents an overview of the current knowledge of these many facets of small GTPase regulation.
Collapse
Affiliation(s)
- Jacqueline Cherfils
- Laboratoire d’Enzymologie et Biochimie Structurales, Centre National de la Recherche Scientifique, Centre deRecherche de Gif, Gif-sur-Yvette, France
| | | |
Collapse
|