1
|
A Avelar R, Gupta R, Carvette G, da Veiga Leprevost F, Jasti M, Colina J, Teitel J, Nesvizhskii AI, O'Connor CM, Hatzoglou M, Shenolikar S, Arvan P, Narla G, DiFeo A. Integrated stress response plasticity governs normal cell adaptation to chronic stress via the PP2A-TFE3-ATF4 pathway. Cell Death Differ 2024; 31:1761-1775. [PMID: 39349971 PMCID: PMC11618521 DOI: 10.1038/s41418-024-01378-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 10/09/2024] Open
Abstract
The integrated stress response (ISR) regulates cell fate during conditions of stress by leveraging the cell's capacity to endure sustainable and efficient adaptive stress responses. Protein phosphatase 2A (PP2A) activity modulation has been shown to be successful in achieving both therapeutic efficacy and safety across various cancer models. However, the molecular mechanisms driving its selective antitumor effects remain unclear. Here, we show for the first time that ISR plasticity relies on PP2A activation to regulate drug response and dictate cellular survival under conditions of chronic stress. We demonstrate that genetic and chemical modulation of the PP2A leads to chronic proteolytic stress and triggers an ISR to dictate whether the cell lives or dies. More specifically, we uncovered that the PP2A-TFE3-ATF4 pathway governs ISR cell plasticity during endoplasmic reticular and cellular stress independent of the unfolded protein response. We further show that normal cells reprogram their genetic signatures to undergo ISR-mediated adaptation and homeostatic recovery thereby avoiding toxicity following PP2A-mediated stress. Conversely, oncogenic specific cytotoxicity induced by chemical modulation of PP2A is achieved by activating chronic and irreversible ISR in cancer cells. Our findings propose that a differential response to chemical modulation of PP2A is determined by intrinsic ISR plasticity, providing a novel biological vulnerability to selectively induce cancer cell death and improve targeted therapeutic efficacy.
Collapse
Affiliation(s)
- Rita A Avelar
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Riya Gupta
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Grace Carvette
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | | | - Medhasri Jasti
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Jose Colina
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Jessica Teitel
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Caitlin M O'Connor
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Maria Hatzoglou
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Shirish Shenolikar
- Duke-NUS Medical School, Singapore, Singapore
- Duke University School of Medicine, Durham, NC, USA
| | - Peter Arvan
- Division of Metabolism Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Goutham Narla
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Analisa DiFeo
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Gangadaran P, Khan F, Rajendran RL, Onkar A, Goenka A, Ahn BC. Unveiling Invisible Extracellular Vesicles: Cutting-Edge Technologies for Their in Vivo Visualization. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2009. [PMID: 39439198 DOI: 10.1002/wnan.2009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs), nanosized lipid bilayer vesicles released by nearly all types of cells, play pivotal roles as intercellular signaling mediators with diverse biological activities. Their adaptability has attracted interest in exploring their role as disease biomarker theranostics. However, the in vivo biodistribution and pharmacokinetic profiles of EVs, particularly following administration into living subjects, remain unclear. Thus, in vivo imaging is vital to enhance our understanding of the homing and retention patterns, blood and tissue half-life, and excretion pathways of exogenous EVs, thereby advancing real-time monitoring within biological systems and their therapeutic applications. This review examines state-of-the-art methods including EV labeling with various agents, including optical imaging, magnetic resonance imaging, and nuclear imaging. The strengths and weaknesses of each technique are comprehensively explored, emphasizing their clinical translation. Despite the potential of EVs as cancer theranostics, achieving a thorough understanding of their in vivo behavior is challenging. This review highlights the urgency of addressing current questions in the biology and therapeutic applications of EVs. It underscores the need for continued research to unravel the complexities surrounding EVs and their potential clinical implications. By identifying these challenges, this review contributes to ongoing efforts to optimize EV imaging techniques for clinical use. Ultimately, bridging the gap between research advancements and clinical applications will facilitate the integration of EV-based theranostics, marking a crucial step toward harnessing the full potential of EVs in medical practice.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Fatima Khan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Akanksha Onkar
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Anshika Goenka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
3
|
Hsiao C, Tsai T, Shen T, Tsai Y, Liao Y, Lee Y, Tsai P. Characterization of a novel TFG variant causing autosomal recessive pure hereditary spastic paraplegia. Ann Clin Transl Neurol 2024; 11:1909-1920. [PMID: 38837630 PMCID: PMC11251477 DOI: 10.1002/acn3.52113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 06/07/2024] Open
Abstract
OBJECTIVE TFG mutations have previously been implicated in autosomal recessive hereditary spastic paraplegia (HSP), also known as SPG57. This study aimed to investigate the clinical and molecular features of TFG mutations in a Taiwanese HSP cohort. METHODS Genetic analysis of TFG was conducted in 242 unrelated Taiwanese HSP patients using a targeted resequencing panel covering the entire coding regions of TFG. Functional assays were performed using an in vitro cell model to assess the impact of TFG variants on protein function. Additionally, other representative TFG mutant proteins were examined to understand the broader implications of TFG mutations in HSP. RESULTS The study identified a novel homozygous TFG c.177A>C (p.(Lys59Asn)) variant in a family with adolescent-onset, pure form HSP. Functional analysis revealed that the Lys59Asn TFG variant, similar to other HSP-associated TFG mutants, exhibited a low affinity between TFG monomers and abnormal assembly of TFG homo-oligomers. These structural alterations led to aberrant intracellular distribution, compromising TFG's protein secretion function and resulting in decreased cellular viability. INTERPRETATION These findings confirm that the homozygous TFG c.177A>C (p.(Lys59Asn)) variant is a novel cause of SPG57. The study expands our understanding of the clinical and mutational spectrum of TFG-associated diseases, highlighting the functional defects associated with this specific TFG variant. Overall, this research contributes to the broader comprehension of the genetic and molecular mechanisms underlying HSP.
Collapse
Affiliation(s)
- Cheng‐Tsung Hsiao
- Department of NeurologyTaipei Veterans General HospitalTaipeiTaiwan
- Department of NeurologyNational Yang Ming Chiao Tung University School of MedicineTaipeiTaiwan
| | - Tzu‐Yun Tsai
- Department of Life SciencesNational Chung Hsing UniversityTaichungTaiwan
| | - Ting‐Yi Shen
- Department of Life SciencesNational Chung Hsing UniversityTaichungTaiwan
| | - Yu‐Shuen Tsai
- Cancer and Immunology Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yi‐Chu Liao
- Department of NeurologyTaipei Veterans General HospitalTaipeiTaiwan
- Department of NeurologyNational Yang Ming Chiao Tung University School of MedicineTaipeiTaiwan
- Brain Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yi‐Chung Lee
- Department of NeurologyTaipei Veterans General HospitalTaipeiTaiwan
- Department of NeurologyNational Yang Ming Chiao Tung University School of MedicineTaipeiTaiwan
- Brain Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Center for Intelligent Drug Systems and Smart Bio‐devices (IDS2B)National Yang Ming Chiao Tung UniversityHsinchuTaiwan
| | - Pei‐Chien Tsai
- Department of Life SciencesNational Chung Hsing UniversityTaichungTaiwan
- The iEGG and Animal Biotechnology Research CenterNational Chung Hsing UniversityTaichungTaiwan
| |
Collapse
|
4
|
Avelar RA, Gupta R, Carvette G, da Veiga Leprevost F, Colina J, Teitel J, Nesvizhskii AI, O’Connor CM, Hatzoglou M, Shenolikar S, Arvan P, Narla G, DiFeo A. Integrated stress response plasticity governs normal cell adaptation to chronic stress via the PP2A-TFE3-ATF4 pathway. RESEARCH SQUARE 2024:rs.3.rs-4013396. [PMID: 38585734 PMCID: PMC10996823 DOI: 10.21203/rs.3.rs-4013396/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The integrated stress response (ISR) regulates cell fate during conditions of stress by leveraging the cell's capacity to endure sustainable and efficient adaptive stress responses. Protein phosphatase 2A (PP2A) activity modulation has been shown to be successful in achieving both therapeutic efficacy and safety across various cancer models; however, the molecular mechanisms driving its selective antitumor effects remain unclear. Here, we show for the first time that ISR plasticity relies on PP2A activation to regulate drug response and dictate cellular fate under conditions of chronic stress. We demonstrate that genetic and chemical modulation of the PP2A leads to chronic proteolytic stress and triggers an ISR to dictate cell fate. More specifically, we uncovered that the PP2A-TFE3-ATF4 pathway governs ISR cell plasticity during endoplasmic reticular and cellular stress independent of the unfolded protein response. We further show that normal cells reprogram their genetic signatures to undergo ISR-mediated adaptation and homeostatic recovery thereby successfully avoiding toxicity following PP2A-mediated stress. Conversely, oncogenic specific cytotoxicity induced by chemical modulation of PP2A is achieved by activating chronic and irreversible ISR in cancer cells. Our findings propose that a differential response to chemical modulation of PP2A is determined by intrinsic ISR plasticity, providing a novel biological vulnerability to selectively induce cancer cell death and improve targeted therapeutic efficacy.
Collapse
Affiliation(s)
- Rita A. Avelar
- Department of Pathology, The University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Riya Gupta
- Department of Pathology, The University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Gracie Carvette
- Department of Pathology, The University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Jose Colina
- Department of Pathology, The University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Jessica Teitel
- Department of Pathology, The University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexey I. Nesvizhskii
- Department of Pathology, The University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Caitlin M. O’Connor
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Genetic Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maria Hatzoglou
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Shirish Shenolikar
- Emeritus Professor, Duke-NUS Medical School, Singapore
- Professor Emeritus, Duke University School of Medicine, USA
| | - Peter Arvan
- Division of Metabolism Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Goutham Narla
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Genetic Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Analisa DiFeo
- Department of Pathology, The University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI 48109, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
5
|
Robinson CM, Duggan A, Forrester A. ER exit in physiology and disease. Front Mol Biosci 2024; 11:1352970. [PMID: 38314136 PMCID: PMC10835805 DOI: 10.3389/fmolb.2024.1352970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/05/2024] [Indexed: 02/06/2024] Open
Abstract
The biosynthetic secretory pathway is comprised of multiple steps, modifications and interactions that form a highly precise pathway of protein trafficking and secretion, that is essential for eukaryotic life. The general outline of this pathway is understood, however the specific mechanisms are still unclear. In the last 15 years there have been vast advancements in technology that enable us to advance our understanding of this complex and subtle pathway. Therefore, based on the strong foundation of work performed over the last 40 years, we can now build another level of understanding, using the new technologies available. The biosynthetic secretory pathway is a high precision process, that involves a number of tightly regulated steps: Protein folding and quality control, cargo selection for Endoplasmic Reticulum (ER) exit, Golgi trafficking, sorting and secretion. When deregulated it causes severe diseases that here we categorise into three main groups of aberrant secretion: decreased, excess and altered secretion. Each of these categories disrupts organ homeostasis differently, effecting extracellular matrix composition, changing signalling events, or damaging the secretory cells due to aberrant intracellular accumulation of secretory proteins. Diseases of aberrant secretion are very common, but despite this, there are few effective therapies. Here we describe ER exit sites (ERES) as key hubs for regulation of the secretory pathway, protein quality control and an integratory hub for signalling within the cell. This review also describes the challenges that will be faced in developing effective therapies, due to the specificity required of potential drug candidates and the crucial need to respect the fine equilibrium of the pathway. The development of novel tools is moving forward, and we can also use these tools to build our understanding of the acute regulation of ERES and protein trafficking. Here we review ERES regulation in context as a therapeutic strategy.
Collapse
Affiliation(s)
- Claire M Robinson
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Aislinn Duggan
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Alison Forrester
- Research Unit of Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| |
Collapse
|
6
|
Coulet M, Lachkar S, Leduc M, Trombe M, Gouveia Z, Perez F, Kepp O, Kroemer G, Basmaciogullari S. Identification of Small Molecules Affecting the Secretion of Therapeutic Antibodies with the Retention Using Selective Hook (RUSH) System. Cells 2023; 12:1642. [PMID: 37371112 DOI: 10.3390/cells12121642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Unlocking cell secretion capacity is of paramount interest for the pharmaceutical industry focused on biologics. Here, we leveraged retention using a selective hook (RUSH) system for the identification of human osteosarcoma U2OS cell secretion modulators, through automated, high-throughput screening of small compound libraries. We created a U2OS cell line which co-expresses a variant of streptavidin addressed to the lumen-facing membrane of the endoplasmic reticulum (ER) and a recombinant anti-PD-L1 antibody. The heavy chain of the antibody was modified at its C-terminus, to which a furin cleavage site, a green fluorescent protein (GFP), and a streptavidin binding peptide (SBP) were added. We show that the U2OS cell line stably expresses the streptavidin hook and the recombinant antibody bait, which is retained in the ER through the streptavidin-SBP interaction. We further document that the addition of biotin to the culture medium triggers the antibody release from the ER, its trafficking through the Golgi where the GFP-SBP moiety is clipped off, and eventually its release in the extra cellular space, with specific antigen-binding properties. The use of this clone in screening campaigns led to the identification of lycorine as a secretion enhancer, and nigericin and tyrphostin AG-879 as secretion inhibitors. Altogether, our data support the utility of this approach for the identification of agents that could be used to improve recombinant production yields and also for a better understanding of the regulatory mechanism at work in the conventional secretion pathway.
Collapse
Affiliation(s)
- Mathilde Coulet
- Sanofi R&D, 94400 Vitry-sur-Seine, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France
| | - Sylvie Lachkar
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France
| | - Marion Leduc
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France
| | | | - Zelia Gouveia
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR 144, 26 rue d'Ulm, 75005 Paris, France
| | - Franck Perez
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR 144, 26 rue d'Ulm, 75005 Paris, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| | | |
Collapse
|
7
|
Sugawara H, Imai J, Yamamoto J, Izumi T, Kawana Y, Endo A, Kohata M, Seike J, Kubo H, Komamura H, Munakata Y, Asai Y, Hosaka S, Sawada S, Kodama S, Takahashi K, Kaneko K, Katagiri H. A highly sensitive strategy for monitoring real-time proliferation of targeted cell types in vivo. Nat Commun 2023; 14:3253. [PMID: 37316473 DOI: 10.1038/s41467-023-38897-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 05/22/2023] [Indexed: 06/16/2023] Open
Abstract
Cell proliferation processes play pivotal roles in timely adaptation to many biological situations. Herein, we establish a highly sensitive and simple strategy by which time-series showing the proliferation of a targeted cell type can be quantitatively monitored in vivo in the same individuals. We generate mice expressing a secreted type of luciferase only in cells producing Cre under the control of the Ki67 promoter. Crossing these with tissue-specific Cre-expressing mice allows us to monitor the proliferation time course of pancreatic β-cells, which are few in number and weakly proliferative, by measuring plasma luciferase activity. Physiological time courses, during obesity development, pregnancy and juvenile growth, as well as diurnal variation, of β-cell proliferation, are clearly detected. Moreover, this strategy can be utilized for highly sensitive ex vivo screening for proliferative factors for targeted cells. Thus, these technologies may contribute to advancements in broad areas of biological and medical research.
Collapse
Affiliation(s)
- Hiroto Sugawara
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junta Imai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Junpei Yamamoto
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohito Izumi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yohei Kawana
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Endo
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masato Kohata
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junro Seike
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Haremaru Kubo
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Komamura
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuichiro Munakata
- Division of Metabolism and Diabetes, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yoichiro Asai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinichiro Hosaka
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shojiro Sawada
- Division of Metabolism and Diabetes, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Shinjiro Kodama
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
8
|
Cruz MA, Gonzalez Y, Vélez Toro JA, Karimzadeh M, Rubbo A, Morris L, Medam R, Splawn T, Archer M, Fernandes RJ, Dennis JE, Kean TJ. Micronutrient optimization for tissue engineered articular cartilage production of type II collagen. Front Bioeng Biotechnol 2023; 11:1179332. [PMID: 37346792 PMCID: PMC10280293 DOI: 10.3389/fbioe.2023.1179332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Tissue Engineering of cartilage has been hampered by the inability of engineered tissue to express native levels of type II collagen in vitro. Inadequate levels of type II collagen are, in part, due to a failure to recapitulate the physiological environment in culture. In this study, we engineered primary rabbit chondrocytes to express a secreted reporter, Gaussia Luciferase, driven by the type II collagen promoter, and applied a Design of Experiments approach to assess chondrogenic differentiation in micronutrient-supplemented medium. Using a Response Surface Model, 240 combinations of micronutrients absent in standard chondrogenic differentiation medium, were screened and assessed for type II collagen promoter-driven Gaussia luciferase expression. While the target of this study was to establish a combination of all micronutrients, alpha-linolenic acid, copper, cobalt, chromium, manganese, molybdenum, vitamins A, E, D and B7 were all found to have a significant effect on type II collagen promoter activity. Five conditions containing all micronutrients predicted to produce the greatest luciferase expression were selected for further study. Validation of these conditions in 3D aggregates identified an optimal condition for type II collagen promoter activity. Engineered cartilage grown in this condition, showed a 170% increase in type II collagen expression (Day 22 Luminescence) and in Young's tensile modulus compared to engineered cartilage in basal media alone.Collagen cross-linking analysis confirmed formation of type II-type II collagen and type II-type IX collagen cross-linked heteropolymeric fibrils, characteristic of mature native cartilage. Combining a Design of Experiments approach and secreted reporter cells in 3D aggregate culture enabled a high-throughput platform that can be used to identify more optimal physiological culture parameters for chondrogenesis.
Collapse
Affiliation(s)
- Maria A. Cruz
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Yamilet Gonzalez
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Javier A. Vélez Toro
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Makan Karimzadeh
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Anthony Rubbo
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Lauren Morris
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Ramapaada Medam
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Taylor Splawn
- Baylor College of Medicine, Houston, TX, United States
| | - Marilyn Archer
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, United States
| | - Russell J. Fernandes
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, United States
| | | | - Thomas J. Kean
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
- Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
9
|
Hall RN, Weill U, Drees L, Leal-Ortiz S, Li H, Khariton M, Chai C, Xue Y, Rosental B, Quake SR, Sánchez Alvarado A, Melosh NA, Fire AZ, Rink JC, Wang B. Heterologous reporter expression in the planarian Schmidtea mediterranea through somatic mRNA transfection. CELL REPORTS METHODS 2022; 2:100298. [PMID: 36313809 PMCID: PMC9606109 DOI: 10.1016/j.crmeth.2022.100298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/11/2022] [Accepted: 08/25/2022] [Indexed: 06/16/2023]
Abstract
Planarians have long been studied for their regenerative abilities. Moving forward, tools for ectopic expression of non-native proteins will be of substantial value. Using a luminescent reporter to overcome the strong autofluorescence of planarian tissues, we demonstrate heterologous protein expression in planarian cells and live animals. Our approach is based on the introduction of mRNA through several nanotechnological and chemical transfection methods. We improve reporter expression by altering untranslated region (UTR) sequences and codon bias, facilitating the measurement of expression kinetics in both isolated cells and whole planarians using luminescence imaging. We also examine protein expression as a function of variations in the UTRs of delivered mRNA, demonstrating a framework to investigate gene regulation at the post-transcriptional level. Together, these advances expand the toolbox for the mechanistic analysis of planarian biology and establish a foundation for the development and expansion of transgenic techniques in this unique model system.
Collapse
Affiliation(s)
| | - Uri Weill
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Leonard Drees
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Sergio Leal-Ortiz
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Hongquan Li
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Margarita Khariton
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Chew Chai
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yuan Xue
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Benyamin Rosental
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Center for Regenerative Medicine and Stem Cells, Ben Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Stephen R. Quake
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Alejandro Sánchez Alvarado
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Howard Hughes Medical Institute, Kansas City, MO 64110, USA
| | - Nicholas A. Melosh
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Andrew Z. Fire
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jochen C. Rink
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Bo Wang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
10
|
Yan Y, Huang J, Huan C, Li L, Li C. Non-Esterified Fatty Acid Induces ER Stress-Mediated Apoptosis via ROS/MAPK Signaling Pathway in Bovine Mammary Epithelial Cells. Metabolites 2022; 12:metabo12090803. [PMID: 36144207 PMCID: PMC9500666 DOI: 10.3390/metabo12090803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 12/03/2022] Open
Abstract
Elevated concentrations of non-esterified fatty acid (NEFA) induced by negative energy balance (NEB) during the transition period of dairy cows is known to be toxic for multiple bovine cell types. However, the effect of NEFA in bovine mammary epithelial cells (BMECs) remains unclear. The present study aimed to explore the role and molecular mechanism of NEFA in endoplasmic reticulum (ER) stress and the subsequent apoptosis in BMECs. The results showed that NEFA increased ER stress and activated the three unfolded protein response (UPR) signaling sub-pathways by upregulating the expression of GRP78, HSP70, XBP1, ATF6, phosphor-PERK, and phosphor-IRE1α. We also found that NEFA dose-dependently induced apoptosis in BMECs, as indicated by flow cytometry analysis and increased apoptotic gene expression. RNA-seq analysis revealed that NEFA induced apoptosis in BMECs, probably via the ATF4-CHOP axis. Mechanistically, our data showed that NEFA increased reactive oxygen species (ROS) levels, resulting in the activation of the MAPK signaling pathway. Moreover, quercetin, a well-known antioxidant, was found to alleviate ER stress-mediated apoptosis in NEFA-treated BMECs. Collectively, our results suggest that NEFA induces ER stress-mediated apoptosis, probably via the ROS/MAPK signaling pathway, as quercetin has been shown to alleviate ER stress-mediated apoptosis in NEFA-treated BMECs.
Collapse
Affiliation(s)
- Yexiao Yan
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Junpeng Huang
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Changchao Huan
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Lian Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chengmin Li
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
- Correspondence:
| |
Collapse
|
11
|
Chien JCY, Tabet E, Pinkham K, da Hora CC, Chang JCY, Lin S, Badr CE, Lai CPK. A multiplexed bioluminescent reporter for sensitive and non-invasive tracking of DNA double strand break repair dynamics in vitro and in vivo. Nucleic Acids Res 2020; 48:e100. [PMID: 32797168 PMCID: PMC7515717 DOI: 10.1093/nar/gkaa669] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/29/2020] [Accepted: 07/31/2020] [Indexed: 12/31/2022] Open
Abstract
Tracking DNA double strand break (DSB) repair is paramount for the understanding and therapeutic development of various diseases including cancers. Herein, we describe a multiplexed bioluminescent repair reporter (BLRR) for non-invasive monitoring of DSB repair pathways in living cells and animals. The BLRR approach employs secreted Gaussia and Vargula luciferases to simultaneously detect homology-directed repair (HDR) and non-homologous end joining (NHEJ), respectively. BLRR data are consistent with next-generation sequencing results for reporting HDR (R2 = 0.9722) and NHEJ (R2 = 0.919) events. Moreover, BLRR analysis allows longitudinal tracking of HDR and NHEJ activities in cells, and enables detection of DSB repairs in xenografted tumours in vivo. Using the BLRR system, we observed a significant difference in the efficiency of CRISPR/Cas9-mediated editing with guide RNAs only 1-10 bp apart. Moreover, BLRR analysis detected altered dynamics for DSB repair induced by small-molecule modulators. Finally, we discovered HDR-suppressing functions of anticancer cardiac glycosides in human glioblastomas and glioma cancer stem-like cells via inhibition of DNA repair protein RAD51 homolog 1 (RAD51). The BLRR method provides a highly sensitive platform to simultaneously and longitudinally track HDR and NHEJ dynamics that is sufficiently versatile for elucidating the physiology and therapeutic development of DSB repair.
Collapse
Affiliation(s)
| | - Elie Tabet
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA.,Department of Biomedical Engineering, University of South Dakota, 4800 N. Career Ave, Suite 221, Sioux Falls, Vermillion, SD 57069, USA
| | - Kelsey Pinkham
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Cintia Carla da Hora
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA.,Neuroscience Program, Harvard Medical School, Boston, MA 02115, USA
| | - Jason Cheng-Yu Chang
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Steven Lin
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Christian E Badr
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA.,Neuroscience Program, Harvard Medical School, Boston, MA 02115, USA
| | - Charles Pin-Kuang Lai
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 10617, Taiwan.,Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan.,Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
12
|
Targeting of HER/ErbB family proteins using broad spectrum Sec61 inhibitors coibamide A and apratoxin A. Biochem Pharmacol 2020; 183:114317. [PMID: 33152346 DOI: 10.1016/j.bcp.2020.114317] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 01/17/2023]
Abstract
Coibamide A is a potent cancer cell toxin and one of a select group of natural products that inhibit protein entry into the secretory pathway via a direct inhibition of the Sec61 protein translocon. Many Sec61 client proteins are clinically relevant drug targets once trafficked to their final destination in or outside the cell, however the use of Sec61 inhibitors to block early biosynthesis of specific proteins is at a pre-clinical stage. In the present study we evaluated the action of coibamide A against human epidermal growth factor receptor (HER, ErbB) proteins in representative breast and lung cancer cell types. HERs were selected for this study as they represent a family of Sec61 clients that is frequently dysregulated in human cancers, including coibamide-sensitive cell types. Although coibamide A inhibits biogenesis of a broad range of Sec61 substrate proteins in a presumed substrate-nonselective manner, endogenous HER3 (ErbB-3) and EGFR (ErbB-1) proteins were more sensitive to coibamide A, and the related Sec61 inhibitor apratoxin A, than HER2 (ErbB-2). Despite this rank order of sensitivity (HER3 > EGFR > HER2), Sec61-dependent inhibition by coibamide A was sufficient to decrease cell surface expression of HER2. We report that coibamide A- or apratoxin A-mediated block of HER3 entry into the secretory pathway is unlikely to be mediated by the HER3 signal peptide alone. HER3 (G11L/S15L), that is fully resistant to the highly substrate-selective cotransin analogue CT8, was more resistant than wild-type HER3 but only at low coibamide A (3 nM) concentrations; HER3 (G11L/S15L) expression was inhibited by higher concentrations of either natural product. Time- and concentration-dependent decreases in HER protein expression induced a commensurate reduction in AKT/MAPK signaling in breast and lung cancer cell types and loss in cell viability. Coibamide A potentiated the cytotoxic efficacy of small molecule kinase inhibitors lapatinib and erlotinib in breast and lung cancer cell types, respectively. These data indicate that natural product modulators of Sec61 function have value as chemical probes to interrogate HER/ErbB signaling in treatment-resistant human cancers.
Collapse
|
13
|
Teng J, Lashgari G, Tabet EI, Tannous BA. The natural compound obtusaquinone targets pediatric high-grade gliomas through ROS-mediated ER stress. Neurooncol Adv 2020; 2:vdaa106. [PMID: 33134921 PMCID: PMC7592425 DOI: 10.1093/noajnl/vdaa106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Pediatric high-grade gliomas (pHGGs) are aggressive primary brain tumors with local invasive growth and poor clinical prognosis. Treatment of pHGGs is particularly challenging given the intrinsic resistance to chemotherapy, an absence of novel therapeutics, and the difficulty of drugs to reach the tumor beds. Accumulating evidence suggests that production of reactive oxygen species (ROS) and misfolded proteins, which typically leads to endoplasmic reticulum (ER) stress, is an essential mechanism in cancer cell survival. Methods Several cell viability assays were used in 6 patient-derived pHGG cultures to evaluate the effect of the natural compound obtusaquinone (OBT) on cytotoxicity. Orthotopic mouse models were used to determine OBT effects in vivo. Immunoblotting, immunostaining, flow cytometry, and biochemical assays were used to investigate the OBT mechanism of action. Results OBT significantly inhibited cell survival of patient-derived pHGG cells in culture. OBT inhibited tumor growth and extended survival in 2 different orthotopic xenograft models. Mechanistically, OBT induced ER stress through abnormal ROS accumulation. Conclusion Our data demonstrate the utility and feasibility of OBT as a potential therapeutic option for improving the clinical treatment of pHGGs.
Collapse
Affiliation(s)
- Jian Teng
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Ghazal Lashgari
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Elie I Tabet
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Sharmin MM, Mizusawa M, Hayashi S, Arai W, Sakata S, Yonekura S. Effects of fatty acids on inducing endoplasmic reticulum stress in bovine mammary epithelial cells. J Dairy Sci 2020; 103:8643-8654. [PMID: 32622599 DOI: 10.3168/jds.2019-18080] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/20/2020] [Indexed: 12/23/2022]
Abstract
Fatty acids play important roles in the regulation of endoplasmic reticulum (ER) stress-induced apoptosis in different cells. Currently, the effects of fatty acids on bovine mammary epithelial cells (MEC) remain unknown. Our study examined bovine MEC viability and measured unfolded protein response (UPR)-related gene and protein expressions following fatty acid treatments. To evaluate the role of fatty acids, we treated MAC-T cells (a line of MEC) with 100 to 400 μM of saturated (palmitic and stearic acid) and unsaturated (palmitoleic, oleic, linoleic, and linolenic acid) fatty acids and 1 to 5 mM of short- and medium-chain fatty acids (acetic, propionic, butyric, and octanoic acid). Thereafter, we determined UPR-related gene expression using quantitative real-time PCR. Palmitic acid stimulated expression of XBP1s, ATF4, ATF6A, and C/EBP homologous protein (CHOP). Stearic acid increased expression of XBP1s and CHOP and decreased expression of ATF4 and ATF6A. Results of Western blot analysis and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay revealed that palmitic and stearic acid reduced MAC-T cell viability and induced extreme ER stress by increasing the protein expression of ER stress markers, such as phospho-PKR-like endoplasmic reticulum kinase, phospho-eIF2α, cleaved CASP-3, and CHOP. Among unsaturated long-chain fatty acids, palmitoleic acid increased expression of ATF4 and ATF6A. Oleic acid increased expression of XBP1s, ATF4, and ATF6A. Linoleic and linolenic acids increased expression of XBP1s, ATF4, and ATF6A but decreased expression of XBP1s and ATF6A at the highest dose. Although palmitoleic, oleic, and linoleic acid decreased CHOP expression, only palmitoleic acid increased MAC-T cell viability. Therefore, unsaturated long-chain fatty acids did not induce severe ER stress. Acetic, propionic, and butyric acids decreased expression of ATF4, ATF6A, and CHOP and increased XBP1s expression. Although only octanoic acid increased ATF4 and ATF6A expressions, it lowered expression of XBP1s and CHOP. Although fatty acid treatment did not increase the levels of ER stress proteins, butyric and octanoic acids reduced cell viability, possibly because of early differentiation. These results suggest that saturated fatty acids play important roles in MEC viability by inducing severe ER stress compared with unsaturated fatty acids. In addition, acetic and propionic acids (short- and medium-chain fatty acids) reduced ER stress. Therefore, the present study reflects the new insight that serum fatty acid concentration plays an important role in maintaining the lactation physiology of dairy cows.
Collapse
Affiliation(s)
- Mst Mamuna Sharmin
- Graduate School of Medicine, Science and Technology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 399-4598, Japan
| | - Moeko Mizusawa
- Graduate School of Science and Technology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 399-4598, Japan
| | - Satoko Hayashi
- Graduate School of Medicine, Science and Technology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 399-4598, Japan
| | - Wataru Arai
- Graduate School of Science and Technology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 399-4598, Japan
| | - Shotaro Sakata
- Graduate School of Science and Technology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 399-4598, Japan
| | - Shinichi Yonekura
- Graduate School of Medicine, Science and Technology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 399-4598, Japan; Graduate School of Science and Technology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 399-4598, Japan; Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 399-4598, Japan.
| |
Collapse
|
15
|
Lopez-Guerrero AM, Espinosa-Bermejo N, Sanchez-Lopez I, Macartney T, Pascual-Caro C, Orantos-Aguilera Y, Rodriguez-Ruiz L, Perez-Oliva AB, Mulero V, Pozo-Guisado E, Martin-Romero FJ. RAC1-Dependent ORAI1 Translocation to the Leading Edge Supports Lamellipodia Formation and Directional Persistence. Sci Rep 2020; 10:6580. [PMID: 32313105 PMCID: PMC7171199 DOI: 10.1038/s41598-020-63353-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/26/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor invasion requires efficient cell migration, which is achieved by the generation of persistent and polarized lamellipodia. The generation of lamellipodia is supported by actin dynamics at the leading edge where a complex of proteins known as the WAVE regulatory complex (WRC) promotes the required assembly of actin filaments to push the front of the cell ahead. By using an U2OS osteosarcoma cell line with high metastatic potential, proven by a xenotransplant in zebrafish larvae, we have studied the role of the plasma membrane Ca2+ channel ORAI1 in this process. We have found that epidermal growth factor (EGF) triggered an enrichment of ORAI1 at the leading edge, where colocalized with cortactin (CTTN) and other members of the WRC, such as CYFIP1 and ARP2/3. ORAI1-CTTN co-precipitation was sensitive to the inhibition of the small GTPase RAC1, an upstream activator of the WRC. RAC1 potentiated ORAI1 translocation to the leading edge, increasing the availability of surface ORAI1 and increasing the plasma membrane ruffling. The role of ORAI1 at the leading edge was studied in genetically engineered U2OS cells lacking ORAI1 expression that helped us to prove the key role of this Ca2+ channel on lamellipodia formation, lamellipodial persistence, and cell directness, which are required for tumor cell invasiveness in vivo.
Collapse
Affiliation(s)
- Aida M Lopez-Guerrero
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Institute of Molecular Pathology Biomarkers, University of Extremadura, Badajoz, 06006, Spain
| | - Noelia Espinosa-Bermejo
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Institute of Molecular Pathology Biomarkers, University of Extremadura, Badajoz, 06006, Spain
| | - Irene Sanchez-Lopez
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Institute of Molecular Pathology Biomarkers, University of Extremadura, Badajoz, 06006, Spain
| | - Thomas Macartney
- MRC- Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, Scotland, United Kingdom
| | - Carlos Pascual-Caro
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Institute of Molecular Pathology Biomarkers, University of Extremadura, Badajoz, 06006, Spain
| | - Yolanda Orantos-Aguilera
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Institute of Molecular Pathology Biomarkers, University of Extremadura, Badajoz, 06006, Spain
| | - Lola Rodriguez-Ruiz
- Department of Cell Biology and Histology, University of Murcia, IMIB-Arrixaca, Murcia, 30100, Spain
| | - Ana B Perez-Oliva
- Department of Cell Biology and Histology, University of Murcia, IMIB-Arrixaca, Murcia, 30100, Spain
| | - Victoriano Mulero
- Department of Cell Biology and Histology, University of Murcia, IMIB-Arrixaca, Murcia, 30100, Spain
| | - Eulalia Pozo-Guisado
- Department of Cell Biology, School of Medicine and Institute of Molecular Pathology Biomarkers, University of Extremadura, Badajoz, 06006, Spain.
| | - Francisco Javier Martin-Romero
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Institute of Molecular Pathology Biomarkers, University of Extremadura, Badajoz, 06006, Spain.
| |
Collapse
|
16
|
Methods for Establishing and Using a Stable Cell Line Expressing Both Gaussia Luciferase and Firefly Luciferase to Screen for Endoplasmic Reticulum Stress. Methods Mol Biol 2020; 2102:531-555. [PMID: 31989575 DOI: 10.1007/978-1-0716-0223-2_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Endoplasmic reticulum (ER) stress is one of the major mechanisms underlying the etiology of multiple diseases and drug-induced toxicity. Gaussia luciferase (Gluc) is a naturally secreted protein that has been used as a reporter for the secretory pathway of ER to enable efficient and real-time monitoring of the ER function. The Gluc assay has been widely used and optimized in various labs. In this chapter, we provide an example of the application of the Gluc assay by establishing a stable cell line expressing both Gluc and firefly luciferase (Fluc) to study ER stress in liver cells. We describe the detailed procedures used in our laboratory for Gluc- and Fluc-containing lentivirus production and titration, for establishing a HepG2-based stable cell line through lentivirus transduction and the validation process. In addition, we provide an example of using the established stable cell line to investigate ER stress.
Collapse
|
17
|
Sicari D, Delaunay‐Moisan A, Combettes L, Chevet E, Igbaria A. A guide to assessing endoplasmic reticulum homeostasis and stress in mammalian systems. FEBS J 2019; 287:27-42. [DOI: 10.1111/febs.15107] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/10/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Daria Sicari
- Inserm U1242 University of Rennes France
- Centre de lutte contre le cancer Eugène Marquis Rennes France
| | - Agnès Delaunay‐Moisan
- Institute for Integrative Biology of the Cell (I2BC) CEA‐Saclay CNRS ISVJC/SBIGEM Laboratoire Stress Oxydant et Cancer Université Paris‐Saclay Gif‐sur‐Yvette France
| | - Laurent Combettes
- UMRS1174 Université Paris Sud Orsay France
- UMRS1174 Institut National de la Santé et de la Recherche Médicale (Inserm) Orsay France
| | - Eric Chevet
- Inserm U1242 University of Rennes France
- Centre de lutte contre le cancer Eugène Marquis Rennes France
| | - Aeid Igbaria
- Inserm U1242 University of Rennes France
- Centre de lutte contre le cancer Eugène Marquis Rennes France
| |
Collapse
|
18
|
Nishiyama T, Kobayashi T, Jirintai S, Kii I, Nagashima S, Prathiwi Primadharsini P, Nishizawa T, Okamoto H. Screening of novel drugs for inhibiting hepatitis E virus replication. J Virol Methods 2019; 270:1-11. [PMID: 31004661 DOI: 10.1016/j.jviromet.2019.04.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 03/05/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022]
Abstract
Hepatitis E, which is caused by hepatitis E virus (HEV), is generally a self-limiting, acute, and rarely fatal disease. It is sometimes fulminant and lethal, especially during pregnancy. Indeed, it occasionally takes a chronic course in immunocompromised individuals. To cure hepatitis E patients, the broad-spectrum antivirals (ribavirin and pegylated interferon α) are used. However, this treatment is insufficient and unsafe in some patients due to embryoteratogenic effects, leukopenia, and thrombocytopenia. In this study, we constructed an HEV replication reporter system with Gaussia luciferase for comprehensively screening anti-HEV drug candidates, and developed a cell-culture system using cells robustly producing HEV to validate the efficacy of anti-HEV drug candidates. We screened anti-HEV drug candidates from United States Food and Drug Administration-approved drugs using the established HEV replication reporter system, and investigated the selected candidates and type III interferons (interferon λ1-3) using the cell-culture system. In conclusion, we constructed an HEV replicon system for anti-HEV drug screening and a novel cell-culture system to strictly evaluate the replication-inhibitory activities of the obtained anti-HEV candidates. Our findings suggested that interferon λ1-3 might be effective for treating hepatitis E.
Collapse
Affiliation(s)
- Takashi Nishiyama
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-Shi, Tochigi 329-0498, Japan
| | - Tominari Kobayashi
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-Shi, Tochigi 329-0498, Japan
| | - Suljid Jirintai
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-Shi, Tochigi 329-0498, Japan; Division of Pathology, Department of Basic Veterinary Medicine, Inner Mongolia Agricultural University College of Medicine, Hohhot, Inner Mongolia, China
| | - Isao Kii
- Common Facilities Unit, Integrated Research Group, Compass to Healthy Life Research Complex Program, RIKEN Cluster for Science, Technology and Innovation Hub, Kobe, Japan
| | - Shigeo Nagashima
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-Shi, Tochigi 329-0498, Japan
| | - Putu Prathiwi Primadharsini
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-Shi, Tochigi 329-0498, Japan
| | - Tsutomu Nishizawa
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-Shi, Tochigi 329-0498, Japan
| | - Hiroaki Okamoto
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-Shi, Tochigi 329-0498, Japan.
| |
Collapse
|
19
|
Simic MS, Moehle EA, Schinzel RT, Lorbeer FK, Halloran JJ, Heydari K, Sanchez M, Jullié D, Hockemeyer D, Dillin A. Transient activation of the UPR ER is an essential step in the acquisition of pluripotency during reprogramming. SCIENCE ADVANCES 2019; 5:eaaw0025. [PMID: 30989118 PMCID: PMC6457941 DOI: 10.1126/sciadv.aaw0025] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/25/2019] [Indexed: 05/11/2023]
Abstract
Somatic cells can be reprogrammed into pluripotent stem cells using the Yamanaka transcription factors. Reprogramming requires both epigenetic landscape reshaping and global remodeling of cell identity, structure, basic metabolic processes, and organelle form and function. We hypothesize that variable regulation of the proteostasis network and its influence upon the protein-folding environment within cells and their organelles is responsible for the low efficiency and stochasticity of reprogramming. We find that the unfolded protein response of the endoplasmic reticulum (UPRER), the mitochondrial UPR, and the heat shock response, which ensure proteome quality during stress, are activated during reprogramming. The UPRER is particularly crucial, and its ectopic, transient activation, genetically or pharmacologically, enhances reprogramming. Last, stochastic activation of the UPRER predicts reprogramming efficiency in naïve cells. Thus, the low efficiency and stochasticity of cellular reprogramming are due partly to the inability to properly initiate the UPRER to remodel the ER and its proteome.
Collapse
Affiliation(s)
- Milos S. Simic
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- California Institute for Regenerative Medicine, Berkeley, CA 94720, USA
- University of California, Berkeley, Berkeley, CA 94720, USA
| | - Erica A. Moehle
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- California Institute for Regenerative Medicine, Berkeley, CA 94720, USA
- University of California, Berkeley, Berkeley, CA 94720, USA
| | - Robert T. Schinzel
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- California Institute for Regenerative Medicine, Berkeley, CA 94720, USA
- University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Jonathan J. Halloran
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- California Institute for Regenerative Medicine, Berkeley, CA 94720, USA
- University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Melissa Sanchez
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- California Institute for Regenerative Medicine, Berkeley, CA 94720, USA
- University of California, Berkeley, Berkeley, CA 94720, USA
| | - Damien Jullié
- University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Andrew Dillin
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- California Institute for Regenerative Medicine, Berkeley, CA 94720, USA
- University of California, Berkeley, Berkeley, CA 94720, USA
- Corresponding author.
| |
Collapse
|
20
|
Zhang Y, Wang C, Gao N, Zhang X, Yu X, Xu J, Gao F. Determination of neutralization activities by a new versatile assay using an HIV-1 genome carrying the Gaussia luciferase gene. J Virol Methods 2019; 267:22-28. [PMID: 30817948 DOI: 10.1016/j.jviromet.2019.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 11/26/2022]
Abstract
Characterization of neutralizing activities are critical to evaluation of the neutralization potency and breadth of monoclonal antibodies or anti-HIV-1 sera elicited during natural HIV-1 infection or by vaccines. We have developed a new neutralization method using the SG3Δenv genome carrying the Gaussia luciferase gene between the env and nef genes. Pseudotype viruses generated using this new SG3Δenv-GLuc backbone together with HIV-1 env genes were infectious to TZM-bl cells, T cell lines and primary T cells. Viral infection can be detected by measuring luciferase activities with both lysed cells and culture supernatants. Neutralization titers in sera from HIV-1-infected individuals against tier 1 and tier 2 viruses were comparable to those determined by the gold standard TZM-bl-firefly method. Since the neutralization activities can be determined by repeatedly measuring luciferase activities in culture supernatants of any cells that are infected by SG3Δenv-GLuc-Env pseudotype viruses, this new method can serve as a versatile and high throughput assay to determine neutralization activities.
Collapse
Affiliation(s)
- Yuepeng Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Chu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Nan Gao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Feng Gao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, 130012, China; Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
21
|
Markova SV, Larionova MD, Vysotski ES. Shining Light on the Secreted Luciferases of Marine Copepods: Current Knowledge and Applications. Photochem Photobiol 2019; 95:705-721. [PMID: 30585639 DOI: 10.1111/php.13077] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 12/18/2018] [Indexed: 01/02/2023]
Abstract
Copepod luciferases-a family of small secretory proteins of 18.4-24.3 kDa, including a signal peptide-are responsible for bright secreted bioluminescence of some marine copepods. The copepod luciferases use coelenterazine as a substrate to produce blue light in a simple oxidation reaction without any additional cofactors. They do not share sequence or structural similarity with other identified bioluminescent proteins including coelenterazine-dependent Renilla and Oplophorus luciferases. The small size, strong luminescence activity and high stability, including thermostability, make secreted copepod luciferases very attractive candidates as reporter proteins which are particularly useful for nondisruptive reporter assays and for high-throughput format. The most known and extensively investigated representatives of this family are the first cloned GpLuc and MLuc luciferases from copepods Gaussia princeps and Metridia longa, respectively. Immediately after cloning, these homologous luciferases were successfully applied as bioluminescent reporters in vivo and in vitro, and since then, the scope of their applications continues to grow. This review is an attempt to systemize and critically evaluate the data scattered through numerous articles regarding the main structural features of copepod luciferases, their luminescent and physicochemical properties. We also review the main trends of their application as bioluminescent reporters in cell and molecular biology.
Collapse
Affiliation(s)
- Svetlana V Markova
- Photobiology Laboratory, Institute of Biophysics SB RAS, Federal Research Center "Krasnoyarsk Science Center SB RAS", Krasnoyarsk, Russia.,Siberian Federal University, Krasnoyarsk, Russia.,N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow, Russia
| | - Marina D Larionova
- Photobiology Laboratory, Institute of Biophysics SB RAS, Federal Research Center "Krasnoyarsk Science Center SB RAS", Krasnoyarsk, Russia.,N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow, Russia
| | - Eugene S Vysotski
- Photobiology Laboratory, Institute of Biophysics SB RAS, Federal Research Center "Krasnoyarsk Science Center SB RAS", Krasnoyarsk, Russia.,N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
22
|
β-arrestin-2 in PAR-1-biased signaling has a crucial role in endothelial function via PDGF-β in stroke. Cell Death Dis 2019; 10:100. [PMID: 30718498 PMCID: PMC6361911 DOI: 10.1038/s41419-019-1375-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 01/07/2019] [Accepted: 01/15/2019] [Indexed: 12/28/2022]
Abstract
Thrombin aggravates ischemic stroke and activated protein C (APC) has a neuroprotective effect. Both proteases interact with protease-activated receptor 1, which exhibits functional selectivity and leads to G-protein- and β-arrestin-mediated-biased signal transduction. We focused on the effect of β-arrestin in PAR-1-biased signaling on endothelial function after stroke or high-fat diet (HFD). Thrombin had a rapid disruptive effect on endothelial function, but APC had a slow protective effect. Paralleled by prolonged MAPK 42/44 signaling activation by APC via β-arrestin-2, a lower cleavage rate of PAR-1 for APC than thrombin was quantitatively visualized by bioluminescence video imaging. HFD-fed mice showed lower β-arrestin-2 levels and more severe ischemic injury. The expression of β-arrestin-2 in capillaries and PDGF-β secretion in HFD-fed mice were reduced in penumbra lesions. These results suggested that β-arrestin-2-MAPK-PDGF-β signaling enhanced protection of endothelial function and barrier integrity after stroke.
Collapse
|
23
|
Jeong YT, Simoneschi D, Keegan S, Melville D, Adler NS, Saraf A, Florens L, Washburn MP, Cavasotto CN, Fenyö D, Cuervo AM, Rossi M, Pagano M. The ULK1-FBXW5-SEC23B nexus controls autophagy. eLife 2018; 7:42253. [PMID: 30596474 PMCID: PMC6351106 DOI: 10.7554/elife.42253] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023] Open
Abstract
In response to nutrient deprivation, the cell mobilizes an extensive amount of membrane to form and grow the autophagosome, allowing the progression of autophagy. By providing membranes and stimulating LC3 lipidation, COPII (Coat Protein Complex II) promotes autophagosome biogenesis. Here, we show that the F-box protein FBXW5 targets SEC23B, a component of COPII, for proteasomal degradation and that this event limits the autophagic flux in the presence of nutrients. In response to starvation, ULK1 phosphorylates SEC23B on Serine 186, preventing the interaction of SEC23B with FBXW5 and, therefore, inhibiting SEC23B degradation. Phosphorylated and stabilized SEC23B associates with SEC24A and SEC24B, but not SEC24C and SEC24D, and they re-localize to the ER-Golgi intermediate compartment, promoting autophagic flux. We propose that, in the presence of nutrients, FBXW5 limits COPII-mediated autophagosome biogenesis. Inhibition of this event by ULK1 ensures efficient execution of the autophagic cascade in response to nutrient starvation.
Collapse
Affiliation(s)
- Yeon-Tae Jeong
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, United States.,Perlmutter NYU Cancer Center, NYU School of Medicine, New York, United States
| | - Daniele Simoneschi
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, United States.,Perlmutter NYU Cancer Center, NYU School of Medicine, New York, United States
| | - Sarah Keegan
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, United States.,Perlmutter NYU Cancer Center, NYU School of Medicine, New York, United States.,Institute for System Genetics, NYU School of Medicine, New York, United States
| | - David Melville
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Natalia S Adler
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina.,Translational Medicine Research Institute (IIMT), CONICET, Facultad de Ciencias Biomédicas and Facultad deIngeniería, Universidad Austral, Pilar-Derqui, Argentina
| | - Anita Saraf
- The Stowers Institute for Medical Research, Kansas, United States
| | - Laurence Florens
- The Stowers Institute for Medical Research, Kansas, United States
| | - Michael P Washburn
- The Stowers Institute for Medical Research, Kansas, United States.,Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas, United States
| | - Claudio N Cavasotto
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina.,Translational Medicine Research Institute (IIMT), CONICET, Facultad de Ciencias Biomédicas and Facultad deIngeniería, Universidad Austral, Pilar-Derqui, Argentina
| | - David Fenyö
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, United States.,Perlmutter NYU Cancer Center, NYU School of Medicine, New York, United States.,Institute for System Genetics, NYU School of Medicine, New York, United States
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, United States
| | - Mario Rossi
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, United States.,Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina.,Perlmutter NYU Cancer Center, NYU School of Medicine, New York, United States
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, United States.,Perlmutter NYU Cancer Center, NYU School of Medicine, New York, United States.,Howard Hughes Medical Institute, New York University School of Medicine, New York, United States
| |
Collapse
|
24
|
Niwa Y, Nakano Y, Suzuki T, Yamagishi M, Otani K, Dohmae N, Simizu S. Topological analysis of DPY19L3, a human C-mannosyltransferase. FEBS J 2018; 285:1162-1174. [PMID: 29405629 DOI: 10.1111/febs.14398] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/19/2018] [Accepted: 01/30/2018] [Indexed: 01/23/2023]
Abstract
C-mannosylation is a rare type of protein glycosylation, the functions and mechanisms of which remain unclear. Recently, we identified DPY19L3 as a C-mannosyltransferase of R-spondin1 in human cells. DPY19L3 is predicted to be a multipass transmembrane protein that localizes to the endoplasmic reticulum (ER); however, its structure is undetermined. In this study, we propose a topological structure of DPY19L3 by in silico analysis and experimental methods such as redox-sensitive luciferase assay and introduction of N-glycosylation sites, suggesting that DPY19L3 comprises 11 transmembrane regions and two re-entrant loops with the N- and C-terminal ends facing the cytoplasm and ER lumen, respectively. Furthermore, DPY19L3 has four predicted N-glycosylation sites, and we have demonstrated that DPY19L3 is N-glycosylated at Asn118 and Asn704 but not Asn319 and Asn439 , supporting our topological model. By mass spectrometry, we measured the C-mannosyltransferase activity of N-glycosylation-defective mutants of DPY19L3 and isoform2, a splice variant, which lacks the C-terminal luminal region of DPY19L3. Isoform2 does not possess C-mannosyltransferase activity, indicating the importance of the C-terminal region; however, N-glycosylations of DPY19L3 do not have any roles for its enzymatic activity. These novel findings on DPY19L3 provide important insights into the mechanism of C-mannosylation.
Collapse
Affiliation(s)
- Yuki Niwa
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Yoshihiko Nakano
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Takehiro Suzuki
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Mizuo Yamagishi
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Kei Otani
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Siro Simizu
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| |
Collapse
|
25
|
Rutkowski DT. Liver function and dysfunction - a unique window into the physiological reach of ER stress and the unfolded protein response. FEBS J 2018; 286:356-378. [PMID: 29360258 DOI: 10.1111/febs.14389] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/08/2018] [Accepted: 01/17/2018] [Indexed: 02/06/2023]
Abstract
The unfolded protein response (UPR) improves endoplasmic reticulum (ER) protein folding in order to alleviate stress. Yet it is becoming increasingly clear that the UPR regulates processes well beyond those directly involved in protein folding, in some cases by mechanisms that fall outside the realm of canonical UPR signaling. These pathways are highly specific from one cell type to another, implying that ER stress signaling affects each tissue in a unique way. Perhaps nowhere is this more evident than in the liver, which-beyond being a highly secretory tissue-is a key regulator of peripheral metabolism and a uniquely proliferative organ upon damage. The liver provides a powerful model system for exploring how and why the UPR extends its reach into physiological processes that occur outside the ER, and how ER stress contributes to the many systemic diseases that involve liver dysfunction. This review will highlight the ways in which the study of ER stress in the liver has expanded the view of the UPR to a response that is a key guardian of cellular homeostasis outside of just the narrow realm of ER protein folding.
Collapse
Affiliation(s)
- D Thomas Rutkowski
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, IA, USA.,Department of Internal Medicine, University of Iowa Carver College of Medicine, IA, USA
| |
Collapse
|
26
|
Martínez G, Duran‐Aniotz C, Cabral‐Miranda F, Vivar JP, Hetz C. Endoplasmic reticulum proteostasis impairment in aging. Aging Cell 2017; 16:615-623. [PMID: 28436203 PMCID: PMC5506418 DOI: 10.1111/acel.12599] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2017] [Indexed: 12/12/2022] Open
Abstract
Perturbed neuronal proteostasis is a salient feature shared by both aging and protein misfolding disorders. The proteostasis network controls the health of the proteome by integrating pathways involved in protein synthesis, folding, trafficking, secretion, and their degradation. A reduction in the buffering capacity of the proteostasis network during aging may increase the risk to undergo neurodegeneration by enhancing the accumulation of misfolded proteins. As almost one-third of the proteome is synthetized at the endoplasmic reticulum (ER), maintenance of its proper function is fundamental to sustain neuronal function. In fact, ER stress is a common feature of most neurodegenerative diseases. The unfolded protein response (UPR) operates as central player to maintain ER homeostasis or the induction of cell death of chronically damaged cells. Here, we discuss recent evidence placing ER stress as a driver of brain aging, and the emerging impact of neuronal UPR in controlling global proteostasis at the whole organismal level. Finally, we discuss possible therapeutic interventions to improve proteostasis and prevent pathological brain aging.
Collapse
Affiliation(s)
- Gabriela Martínez
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
- Center for Integrative BiologyUniversidad MayorSantiagoChile
| | - Claudia Duran‐Aniotz
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
| | - Felipe Cabral‐Miranda
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
- Instituto de Ciências BiomédicasUniversidade Federal do Rio de JaneiroRio de JaneiroBrasil
| | - Juan P. Vivar
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
| | - Claudio Hetz
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
- Buck Institute for Research on AgingNovatoCA94945USA
- Department of Immunology and Infectious diseasesHarvard School of Public HealthBostonMA02115USA
| |
Collapse
|
27
|
Ignashkova TI, Gendarme M, Peschk K, Eggenweiler HM, Lindemann RK, Reiling JH. Cell survival and protein secretion associated with Golgi integrity in response to Golgi stress-inducing agents. Traffic 2017; 18:530-544. [PMID: 28485883 DOI: 10.1111/tra.12493] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/05/2017] [Accepted: 05/05/2017] [Indexed: 12/29/2022]
Abstract
The Golgi apparatus is part of the secretory pathway and of central importance for modification, transport and sorting of proteins and lipids. ADP-ribosylation factors, whose activation can be blocked by brefeldin A (BFA), play a major role in functioning of the Golgi network and regulation of membrane traffic and are also involved in proliferation and migration of cancer cells. Due to high cytotoxicity and poor bioavailability, BFA has not passed the preclinical stage of drug development. Recently, AMF-26 and golgicide A have been described as novel inhibitors of the Golgi system with antitumor or bactericidal properties. We provide here further evidence that AMF-26 closely mirrors the mode of action of BFA but is less potent. Using several human cancer cell lines, we studied the effects of AMF-26, BFA and golgicide A on cell homeostasis including Golgi structure, endoplasmic reticulum (ER) stress markers, secretion and viability, and found overall a significant correlation between these parameters. Furthermore, modulation of ADP-ribosylation factor expression has a profound impact on Golgi organization and survival in response to Golgi stress inducers.
Collapse
Affiliation(s)
- Tatiana I Ignashkova
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Heidelberg, Germany
| | - Mathieu Gendarme
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Heidelberg, Germany
| | - Katrin Peschk
- Medicinal Chemistry, Merck Biopharma, Merck KGaA, Darmstadt, Germany
| | | | - Ralph K Lindemann
- Translational Innovation Platform Oncology, Merck Biopharma, Merck KGaA, Darmstadt, Germany
| | - Jan H Reiling
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Heidelberg, Germany
| |
Collapse
|
28
|
Je HJ, Kim MG, Kwon HJ. Bioluminescence Assays for Monitoring Chondrogenic Differentiation and Cartilage Regeneration. SENSORS 2017; 17:s17061306. [PMID: 28587284 PMCID: PMC5492100 DOI: 10.3390/s17061306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 05/24/2017] [Accepted: 06/02/2017] [Indexed: 02/06/2023]
Abstract
Since articular cartilage has a limited regeneration potential, for developing biological therapies for cartilage regeneration it is important to study the mechanisms underlying chondrogenesis of stem cells. Bioluminescence assays can visualize a wide range of biological phenomena such as gene expression, signaling, metabolism, development, cellular movements, and molecular interactions by using visible light and thus contribute substantially to elucidation of their biological functions. This article gives a concise review to introduce basic principles of bioluminescence assays and applications of the technology to visualize the processes of chondrogenesis and cartilage regeneration. Applications of bioluminescence assays have been highlighted in the methods of real-time monitoring of gene expression and intracellular levels of biomolecules and noninvasive cell tracking within animal models. This review suggests that bioluminescence assays can be applied towards a visual understanding of chondrogenesis and cartilage regeneration.
Collapse
Affiliation(s)
- Hyeon Jeong Je
- Department of Physical Therapy and Rehabilitation Science, College of Health Science, Eulji University, Gyeonggi 13135, Korea.
| | - Min Gu Kim
- Department of Physical Therapy and Rehabilitation Science, College of Health Science, Eulji University, Gyeonggi 13135, Korea.
| | - Hyuck Joon Kwon
- Department of Physical Therapy and Rehabilitation Science, College of Health Science, Eulji University, Gyeonggi 13135, Korea.
| |
Collapse
|
29
|
Zhang Z, Stickney Z, Duong N, Curley K, Lu B. AAV-based dual-reporter circuit for monitoring cell signaling in living human cells. J Biol Eng 2017; 11:18. [PMID: 28592991 PMCID: PMC5458475 DOI: 10.1186/s13036-017-0060-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/20/2017] [Indexed: 01/12/2023] Open
Abstract
Background High-throughput methods based on molecular reporters have greatly advanced our knowledge of cell signaling in mammalian cells. However, their ability to monitor various types of cells is markedly limited by the inefficiency of reporter gene delivery. Recombinant adeno-associated virus (AAV) vectors are efficient tools widely used for delivering and expressing transgenes in diverse animal cells in vitro and in vivo. Here we present the design, construction and validation of a novel AAV-based dual-reporter circuit that can be used to monitor and quantify cell signaling in living human cells. Results We first design and construct the AAV-based reporter system. We then validate the versatility and specificity of this system in monitoring and quantifying two important cell signaling pathways, inflammation (NFκB) and cell growth and differentiation (AP-1), in cultured HEK293 and MCF-7 cells. Our results demonstrate that the AAV reporter system is both specific and versatile, and it can be used in two common experimental protocols including transfection with plasmid DNA and transduction with packaged viruses. Importantly, this system is efficient, with a high signal-to-background noise ratio, and can be easily adapted to monitor other common signaling pathways. Conclusions The AAV-based system extends the dual-reporter technology to more cell types, allowing for cost-effective and high throughput applications. Electronic supplementary material The online version of this article (doi:10.1186/s13036-017-0060-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhiwen Zhang
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053 USA
| | - Zachary Stickney
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053 USA
| | - Natalie Duong
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053 USA
| | - Kevin Curley
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053 USA
| | - Biao Lu
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053 USA
| |
Collapse
|
30
|
Ramírez-Peinado S, Ignashkova TI, van Raam BJ, Baumann J, Sennott EL, Gendarme M, Lindemann RK, Starnbach MN, Reiling JH. TRAPPC13 modulates autophagy and the response to Golgi stress. J Cell Sci 2017; 130:2251-2265. [PMID: 28536105 DOI: 10.1242/jcs.199521] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 05/22/2017] [Indexed: 01/05/2023] Open
Abstract
Tether complexes play important roles in endocytic and exocytic trafficking of lipids and proteins. In yeast, the multisubunit transport protein particle (TRAPP) tether regulates endoplasmic reticulum (ER)-to-Golgi and intra-Golgi transport and is also implicated in autophagy. In addition, the TRAPP complex acts as a guanine nucleotide exchange factor (GEF) for Ypt1, which is homologous to human Rab1a and Rab1b. Here, we show that human TRAPPC13 and other TRAPP subunits are critically involved in the survival response to several Golgi-disrupting agents. Loss of TRAPPC13 partially preserves the secretory pathway and viability in response to brefeldin A, in a manner that is dependent on ARF1 and the large GEF GBF1, and concomitant with reduced caspase activation and ER stress marker induction. TRAPPC13 depletion reduces Rab1a and Rab1b activity, impairs autophagy and leads to increased infectivity to the pathogenic bacterium Shigella flexneri in response to brefeldin A. Thus, our results lend support for the existence of a mammalian TRAPPIII complex containing TRAPPC13, which is important for autophagic flux under certain stress conditions.
Collapse
Affiliation(s)
- Silvia Ramírez-Peinado
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Im Neuenheimer Feld 583, Heidelberg 69120, Germany
| | - Tatiana I Ignashkova
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Im Neuenheimer Feld 583, Heidelberg 69120, Germany
| | - Bram J van Raam
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Im Neuenheimer Feld 583, Heidelberg 69120, Germany
| | - Jan Baumann
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Im Neuenheimer Feld 583, Heidelberg 69120, Germany
| | - Erica L Sennott
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Mathieu Gendarme
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Im Neuenheimer Feld 583, Heidelberg 69120, Germany
| | - Ralph K Lindemann
- Merck Serono TA Oncology, Merck KGaA, Frankfurter Str. 250, Darmstadt D-64293, Germany
| | - Michael N Starnbach
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jan H Reiling
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Im Neuenheimer Feld 583, Heidelberg 69120, Germany
| |
Collapse
|
31
|
Quantitative visualization of synchronized insulin secretion from 3D-cultured cells. Biochem Biophys Res Commun 2017; 486:886-892. [PMID: 28342877 DOI: 10.1016/j.bbrc.2017.03.105] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 11/20/2022]
Abstract
Quantitative visualization of synchronized insulin secretion was performed in an isolated rat pancreatic islet and a spheroid of rat pancreatic beta cell line using a method of video-rate bioluminescence imaging. Video-rate images of insulin secretion from 3D-cultured cells were obtained by expressing the fusion protein of insulin and Gaussia luciferase (Insulin-GLase). A subclonal rat INS-1E cell line stably expressing Insulin-GLase, named iGL, was established and a cluster of iGL cells showed oscillatory insulin secretion that was completely synchronized in response to high glucose. Furthermore, we demonstrated the effect of an antidiabetic drug, glibenclamide, on synchronized insulin secretion from 2D- and 3D-cultured iGL cells. The amount of secreted Insulin-GLase from iGL cells was also determined by a luminometer. Thus, our bioluminescence imaging method could generally be used for investigating protein secretion from living 3D-cultured cells. In addition, iGL cell line would be valuable for evaluating antidiabetic drugs.
Collapse
|
32
|
Russo J, Lee JE, López CM, Anderson J, Nguyen TMP, Heck AM, Wilusz J, Wilusz CJ. The CELF1 RNA-Binding Protein Regulates Decay of Signal Recognition Particle mRNAs and Limits Secretion in Mouse Myoblasts. PLoS One 2017; 12:e0170680. [PMID: 28129347 PMCID: PMC5271678 DOI: 10.1371/journal.pone.0170680] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/09/2017] [Indexed: 02/06/2023] Open
Abstract
We previously identified several mRNAs encoding components of the secretory pathway, including signal recognition particle (SRP) subunit mRNAs, among transcripts associated with the RNA-binding protein CELF1. Through immunoprecipitation of RNAs crosslinked to CELF1 in myoblasts and in vitro binding assays using recombinant CELF1, we now provide evidence that CELF1 directly binds the mRNAs encoding each of the subunits of the SRP. Furthermore, we determined the half-lives of the Srp transcripts in control and CELF1 knockdown myoblasts. Our results indicate CELF1 is a destabilizer of at least five of the six Srp transcripts and that the relative abundance of the SRP proteins is out of balance when CELF1 is depleted. CELF1 knockdown myoblasts exhibit altered secretion of a luciferase reporter protein and are impaired in their ability to migrate and close a wound, consistent with a defect in the secreted extracellular matrix. Importantly, similar defects in wound healing are observed when SRP subunit imbalance is induced by over-expression of SRP68. Our studies support the existence of an RNA regulon containing Srp mRNAs that is controlled by CELF1. One implication is that altered function of CELF1 in myotonic dystrophy may contribute to changes in the extracellular matrix of affected muscle through defects in secretion.
Collapse
Affiliation(s)
- Joseph Russo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jerome E. Lee
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- Program in Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Carolina M. López
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - John Anderson
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Thuy-mi P. Nguyen
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Adam M. Heck
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- Program in Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jeffrey Wilusz
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- Program in Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Carol J. Wilusz
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- Program in Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
33
|
Abstract
Secreted reporter proteins are reliable modalities for monitoring of different biological processes, which can be measured longitudinally in conditioned medium of cultured cells or body fluids such as blood and urine, ex vivo. In this chapter, we will explore established secreted reporters and their applications and limitations for monitoring of promoter function. We will also describe both cell-based and blood-based assays for detecting three commonly used reporters: secreted alkaline phosphatase (SEAP ), Gaussia luciferase (Gluc), and Vargula luciferase (Vluc).
Collapse
Affiliation(s)
- Ghazal Lashgari
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Rami S Kantar
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA.
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA.
- Neuroscience Center, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, MA, 02129, USA.
| |
Collapse
|
34
|
Zadoo S, Nguyen A, Zode G, Hulleman JD. A Novel Luciferase Assay For Sensitively Monitoring Myocilin Variants in Cell Culture. Invest Ophthalmol Vis Sci 2016; 57:1939-50. [PMID: 27092720 PMCID: PMC5110263 DOI: 10.1167/iovs.15-18789] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose Primary open angle glaucoma–associated mutations in myocilin (MYOC) cause protein “nonsecretion,” rendering secreted MYOC difficult to detect using conventional techniques. This study focused on developing and using an assay that can quickly and easily detect mutant MYOC secretion. Methods We fused Gaussia luciferase (eGLuc2) to MYOC variants and expressed the constructs in HEK-293T and NTM-5 cells. Secreted and intracellular levels of MYOC eGLuc2 variants were evaluated by Western blotting and compared to untagged and FLAG-tagged MYOC constructs. Secreted and soluble intracellular MYOC eGLuc2 were measured by a GLuc assay. The secretion of nine additional MYOC mutants was assayed in conditioned media from transfected cells to test the applicability of the assay for monitoring other MYOC variants. Results Myocilin eGLuc2 behaved similarly to untagged and FLAG-tagged MYOC with respect to secretion, soluble intracellular levels, and in response to drug treatment. The GLuc assay could sensitively detect Y437H MYOC secretion 30 minutes after media change. Gaussia luciferase fused variants followed anticipated trends; nonpathogenic variants (D208E, G244V) were secreted at wild-type (WT) levels, whereas predicted disease-causing variants (C245Y, G246R, E300K, Y437H, I477N) demonstrated substantial secretion defects. Secretion defects caused by the C245Y, G246R, and Y437H mutations were partially rescued by permissive growth temperature. Interestingly, however, this increase in secretion was independent of newly synthesized protein. Conclusions Fusion of eGLuc2 to MYOC does not significantly change the behavior of MYOC. This newly developed MYOC reporter system can be used to study engineered MYOC variants and potentially to identify modulators of MYOC secretion and function.
Collapse
Affiliation(s)
- Serena Zadoo
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Annie Nguyen
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Gulab Zode
- Department of Cell Biology & Immunology and the North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - John D Hulleman
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States 3Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
35
|
Alessandrini F, Ceresa D, Appolloni I, Marubbi D, Malatesta P. Noninvasive Monitoring of Glioma Growth in the Mouse. J Cancer 2016; 7:1791-1797. [PMID: 27698917 PMCID: PMC5039361 DOI: 10.7150/jca.15564] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/17/2016] [Indexed: 02/01/2023] Open
Abstract
Malignant gliomas are the most common and deadly primary malignant brain tumors. In vivo orthotopic models could doubtless represent an appropriate tool to test novel treatment for gliomas. However, methods commonly used to monitor the growth of glioma inside the mouse brain are time consuming and invasive. We tested the reliability of a minimally invasive procedure, based on a secreted luciferase (Gaussia luciferase), to frequently monitor the changes of glioma size. Gluc activity was evaluated from blood samples collected from the tail tip of mice twice a week, allowing to make a growth curve for the tumors. We validated the correlation between Gluc activity and tumor size by analysing the tumor after brain dissection. We found that this method is reliable for monitoring human glioma transplanted in immunodeficient mice, but it has strong limitation in immunocompetent models, where an immune response against the luciferase is developed during the first weeks after transplant.
Collapse
Affiliation(s)
- Francesco Alessandrini
- Department of Experimental Medicine (DiMES), University of Genoa, Leon Battista Alberti 2, 16132, Genoa-Italy
| | - Davide Ceresa
- Department of Experimental Medicine (DiMES), University of Genoa, Leon Battista Alberti 2, 16132, Genoa-Italy
| | - Irene Appolloni
- IRCCS-AOU San Martino-IST, Largo Rosanna Benzi 10, 16132, Genoa-Italy
| | - Daniela Marubbi
- Department of Experimental Medicine (DiMES), University of Genoa, Leon Battista Alberti 2, 16132, Genoa-Italy.; IRCCS-AOU San Martino-IST, Largo Rosanna Benzi 10, 16132, Genoa-Italy
| | - Paolo Malatesta
- Department of Experimental Medicine (DiMES), University of Genoa, Leon Battista Alberti 2, 16132, Genoa-Italy.; IRCCS-AOU San Martino-IST, Largo Rosanna Benzi 10, 16132, Genoa-Italy
| |
Collapse
|
36
|
Dupuis N, Fafouri A, Bayot A, Kumar M, Lecharpentier T, Ball G, Edwards D, Bernard V, Dournaud P, Drunat S, Vermelle-Andrzejewski M, Vilain C, Abramowicz M, Désir J, Bonaventure J, Gareil N, Boncompain G, Csaba Z, Perez F, Passemard S, Gressens P, El Ghouzzi V. Dymeclin deficiency causes postnatal microcephaly, hypomyelination and reticulum-to-Golgi trafficking defects in mice and humans. Hum Mol Genet 2015; 24:2771-83. [PMID: 25652408 DOI: 10.1093/hmg/ddv038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 01/31/2015] [Indexed: 01/02/2023] Open
Abstract
Dymeclin is a Golgi-associated protein whose deficiency causes Dyggve-Melchior-Clausen syndrome (DMC, MIM #223800), a rare recessively inherited spondyloepimetaphyseal dysplasia consistently associated with postnatal microcephaly and intellectual disability. While the skeletal phenotype of DMC patients has been extensively described, very little is known about their cerebral anomalies, which result in brain growth defects and cognitive dysfunction. We used Dymeclin-deficient mice to determine the cause of microcephaly and to identify defective mechanisms at the cellular level. Brain weight and volume were reduced in all mutant mice from postnatal day 5 onward. Mutant mice displayed a narrowing of the frontal cortex, although cortical layers were normally organized. Interestingly, the corpus callosum was markedly thinner, a characteristic we also identified in DMC patients. Consistent with this, the myelin sheath was thinner, less compact and not properly rolled, while the number of mature oligodendrocytes and their ability to produce myelin basic protein were significantly decreased. Finally, cortical neurons from mutant mice and primary fibroblasts from DMC patients displayed substantially delayed endoplasmic reticulum to Golgi trafficking, which could be fully rescued upon Dymeclin re-expression. These findings indicate that Dymeclin is crucial for proper myelination and anterograde neuronal trafficking, two processes that are highly active during postnatal brain maturation.
Collapse
Affiliation(s)
- Nina Dupuis
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France
| | - Assia Fafouri
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France
| | - Aurélien Bayot
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France
| | - Manoj Kumar
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France
| | - Tifenn Lecharpentier
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France
| | - Gareth Ball
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - David Edwards
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Véronique Bernard
- CNRS UMR7224, Inserm, U952, Paris, France, Univ Pierre et Marie Curie, Paris, France
| | - Pascal Dournaud
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France
| | - Séverine Drunat
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France, Service de Génétique Clinique, AP-HP, Hôpital Robert Debré, Paris, France
| | | | - Catheline Vilain
- Medical Genetics Department, Hôpital Erasme, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marc Abramowicz
- Medical Genetics Department, Hôpital Erasme, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Julie Désir
- Medical Genetics Department, Hôpital Erasme, Université Libre de Bruxelles (ULB), Brussels, Belgium, Institut de Pathologie et de Génétique, Gosselies, Belgium
| | - Jacky Bonaventure
- CNRS UMR3347, Orsay, France, Institut Curie, Centre de Recherche, Paris, France
| | - Nelly Gareil
- CNRS UMR144, Paris, France and Institut Curie, Centre de Recherche, Paris, France
| | - Gaelle Boncompain
- CNRS UMR144, Paris, France and Institut Curie, Centre de Recherche, Paris, France
| | - Zsolt Csaba
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France
| | - Franck Perez
- CNRS UMR144, Paris, France and Institut Curie, Centre de Recherche, Paris, France
| | - Sandrine Passemard
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France, Service de Génétique Clinique, AP-HP, Hôpital Robert Debré, Paris, France
| | - Pierre Gressens
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France, Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Vincent El Ghouzzi
- Inserm, U1141, Paris, France, Sorbonne Paris Cité, Univ Paris Diderot, UMRS 1141, Paris, France,
| |
Collapse
|
37
|
Biological production of an integrin αvβ3 targeting imaging probe and functional verification. BIOMED RESEARCH INTERNATIONAL 2015; 2015:681012. [PMID: 25654118 PMCID: PMC4310313 DOI: 10.1155/2015/681012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/30/2014] [Accepted: 10/30/2014] [Indexed: 11/17/2022]
Abstract
The aim of the present study is to establish a bacterial clone capable of secreting an integrin αvβ3 targeting probe with bioluminescent and fluorescent activities, and to verify its specific targeting and optical activities using molecular imaging. A bacterial vector expressing a fusion of secretory Gaussia luciferase (sGluc), mCherry, and RGD (sGluc-mCherry-RGDX3; GCR), and a control vector expressing a fusion of secretory Gaussia luciferase and mCherry (sGluc-mCherry; GC) were constructed. The GCR and GC proteins were expressed in E. coli and secreted into the growth medium, which showed an approximately 10-fold higher luciferase activity than the bacterial lysate. Successful purification of GCR and GC was achieved using the 6X His-tag method. The GCR protein bound with higher affinity to U87MG cells than CHO cells in confocal microscopy and IVIS imaging, and also showed a high affinity for integrin αvβ3 expressing tumor xenografts in an in vivo animal model. An E. coli clone was established to secrete an integrin αvβ3 targeting imaging probe with bioluminescent and fluorescent activities. The probe was produced feasibly and at low cost, and has shown to be useful for the assessment of angiogenesis in vitro and in vivo.
Collapse
|
38
|
Synofzik M, Haack T, Kopajtich R, Gorza M, Rapaport D, Greiner M, Schönfeld C, Freiberg C, Schorr S, Holl R, Gonzalez M, Fritsche A, Fallier-Becker P, Zimmermann R, Strom T, Meitinger T, Züchner S, Schüle R, Schöls L, Prokisch H. Absence of BiP co-chaperone DNAJC3 causes diabetes mellitus and multisystemic neurodegeneration. Am J Hum Genet 2014; 95:689-97. [PMID: 25466870 DOI: 10.1016/j.ajhg.2014.10.013] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/28/2014] [Indexed: 10/24/2022] Open
Abstract
Diabetes mellitus and neurodegeneration are common diseases for which shared genetic factors are still only partly known. Here, we show that loss of the BiP (immunoglobulin heavy-chain binding protein) co-chaperone DNAJC3 leads to diabetes mellitus and widespread neurodegeneration. We investigated three siblings with juvenile-onset diabetes and central and peripheral neurodegeneration, including ataxia, upper-motor-neuron damage, peripheral neuropathy, hearing loss, and cerebral atrophy. Exome sequencing identified a homozygous stop mutation in DNAJC3. Screening of a diabetes database with 226,194 individuals yielded eight phenotypically similar individuals and one family carrying a homozygous DNAJC3 deletion. DNAJC3 was absent in fibroblasts from all affected subjects in both families. To delineate the phenotypic and mutational spectrum and the genetic variability of DNAJC3, we analyzed 8,603 exomes, including 506 from families affected by diabetes, ataxia, upper-motor-neuron damage, peripheral neuropathy, or hearing loss. This analysis revealed only one further loss-of-function allele in DNAJC3 and no further associations in subjects with only a subset of the features of the main phenotype. Our findings demonstrate that loss-of-function DNAJC3 mutations lead to a monogenic, recessive form of diabetes mellitus in humans. Moreover, they present a common denominator for diabetes and widespread neurodegeneration. This complements findings from mice in which knockout of Dnajc3 leads to diabetes and modifies disease in a neurodegenerative model of Marinesco-Sjögren syndrome.
Collapse
|
39
|
Kim JE, Kalimuthu S, Ahn BC. In vivo cell tracking with bioluminescence imaging. Nucl Med Mol Imaging 2014; 49:3-10. [PMID: 25774232 DOI: 10.1007/s13139-014-0309-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 11/06/2014] [Accepted: 11/11/2014] [Indexed: 12/29/2022] Open
Abstract
Molecular imaging is a fast growing biomedical research that allows the visual representation, characterization and quantification of biological processes at the cellular and subcellular levels within intact living organisms. In vivo tracking of cells is an indispensable technology for development and optimization of cell therapy for replacement or renewal of damaged or diseased tissue using transplanted cells, often autologous cells. With outstanding advantages of bioluminescence imaging, the imaging approach is most commonly applied for in vivo monitoring of transplanted stem cells or immune cells in order to assess viability of administered cells with therapeutic efficacy in preclinical small animal models. In this review, a general overview of bioluminescence is provided and recent updates of in vivo cell tracking using the bioluminescence signal are discussed.
Collapse
Affiliation(s)
- Jung Eun Kim
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk 2-ga, Jung Gu, Daegu, Republic of Korea 700-721
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk 2-ga, Jung Gu, Daegu, Republic of Korea 700-721
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk 2-ga, Jung Gu, Daegu, Republic of Korea 700-721
| |
Collapse
|
40
|
Roth DM, Hutt DM, Tong J, Bouchecareilh M, Wang N, Seeley T, Dekkers JF, Beekman JM, Garza D, Drew L, Masliah E, Morimoto RI, Balch WE. Modulation of the maladaptive stress response to manage diseases of protein folding. PLoS Biol 2014; 12:e1001998. [PMID: 25406061 PMCID: PMC4236052 DOI: 10.1371/journal.pbio.1001998] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 10/07/2014] [Indexed: 12/31/2022] Open
Abstract
Diseases of protein folding arise because of the inability of an altered peptide sequence to properly engage protein homeostasis components that direct protein folding and function. To identify global principles of misfolding disease pathology we examined the impact of the local folding environment in alpha-1-antitrypsin deficiency (AATD), Niemann-Pick type C1 disease (NPC1), Alzheimer's disease (AD), and cystic fibrosis (CF). Using distinct models, including patient-derived cell lines and primary epithelium, mouse brain tissue, and Caenorhabditis elegans, we found that chronic expression of misfolded proteins not only triggers the sustained activation of the heat shock response (HSR) pathway, but that this sustained activation is maladaptive. In diseased cells, maladaptation alters protein structure-function relationships, impacts protein folding in the cytosol, and further exacerbates the disease state. We show that down-regulation of this maladaptive stress response (MSR), through silencing of HSF1, the master regulator of the HSR, restores cellular protein folding and improves the disease phenotype. We propose that restoration of a more physiological proteostatic environment will strongly impact the management and progression of loss-of-function and gain-of-toxic-function phenotypes common in human disease.
Collapse
Affiliation(s)
- Daniela Martino Roth
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Darren M. Hutt
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jiansong Tong
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Marion Bouchecareilh
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Ning Wang
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois, United States of America
| | - Theo Seeley
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Johanna F. Dekkers
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
- Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
| | - Jeffrey M. Beekman
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
- Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
| | - Dan Garza
- Proteostasis Therapeutics Inc., Cambridge, Massachusetts, United States of America
| | - Lawrence Drew
- Proteostasis Therapeutics Inc., Cambridge, Massachusetts, United States of America
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, California, United States of America
| | - Richard I. Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois, United States of America
| | - William E. Balch
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
- The Institute for Childhood and Neglected Diseases, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
Shah FA, Fatima K, Sabir S, Ali S, Fischer A, Qadri MI. Design, synthesis, and Gaussia luciferase Assay of triorganotin(IV)-based HCV inhibitors. Med Chem Res 2014. [DOI: 10.1007/s00044-014-1242-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
42
|
Unfolded protein response activation reduces secretion and extracellular aggregation of amyloidogenic immunoglobulin light chain. Proc Natl Acad Sci U S A 2014; 111:13046-51. [PMID: 25157167 DOI: 10.1073/pnas.1406050111] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Light-chain amyloidosis (AL) is a degenerative disease characterized by the extracellular aggregation of a destabilized amyloidogenic Ig light chain (LC) secreted from a clonally expanded plasma cell. Current treatments for AL revolve around ablating the cancer plasma cell population using chemotherapy regimens. Unfortunately, this approach is limited to the ∼ 70% of patients who do not exhibit significant organ proteotoxicity and can tolerate chemotherapy. Thus, identifying new therapeutic strategies to alleviate LC organ proteotoxicity should allow AL patients with significant cardiac and/or renal involvement to subsequently tolerate established chemotherapy treatments. Using a small-molecule screening approach, the unfolded protein response (UPR) was identified as a cellular signaling pathway whose activation selectively attenuates secretion of amyloidogenic LC, while not affecting secretion of a nonamyloidogenic LC. Activation of the UPR-associated transcription factors XBP1s and/or ATF6 in the absence of stress recapitulates the selective decrease in amyloidogenic LC secretion by remodeling the endoplasmic reticulum proteostasis network. Stress-independent activation of XBP1s, or especially ATF6, also attenuates extracellular aggregation of amyloidogenic LC into soluble aggregates. Collectively, our results show that stress-independent activation of these adaptive UPR transcription factors offers a therapeutic strategy to reduce proteotoxicity associated with LC aggregation.
Collapse
|
43
|
Hettich J, Ryan SD, de Souza ON, Saraiva Macedo Timmers LF, Tsai S, Atai NA, da Hora CC, Zhang X, Kothary R, Snapp E, Ericsson M, Grundmann K, Breakefield XO, Nery FC. Biochemical and cellular analysis of human variants of the DYT1 dystonia protein, TorsinA/TOR1A. Hum Mutat 2014; 35:1101-13. [PMID: 24930953 DOI: 10.1002/humu.22602] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 06/04/2014] [Indexed: 12/24/2022]
Abstract
Early-onset dystonia is associated with the deletion of one of a pair of glutamic acid residues (c.904_906delGAG/c.907_909delGAG; p.Glu302del/Glu303del; ΔE 302/303) near the carboxyl-terminus of torsinA, a member of the AAA(+) protein family that localizes to the endoplasmic reticulum lumen and nuclear envelope. This deletion commonly underlies early-onset DYT1 dystonia. While the role of the disease-causing mutation, torsinAΔE, has been established through genetic association studies, it is much less clear whether other rare human variants of torsinA are pathogenic. Two missense variations have been described in single patients: R288Q (c.863G>A; p.Arg288Gln; R288Q) identified in a patient with onset of severe generalized dystonia and myoclonus since infancy and F205I (c.613T>A, p.Phe205Ile; F205I) in a psychiatric patient with late-onset focal dystonia. In this study, we have undertaken a series of analyses comparing the biochemical and cellular effects of these rare variants to torsinAΔE and wild-type (wt) torsinA to reveal whether there are common dysfunctional features. The results revealed that the variants, R288Q and F205I, are more similar in their properties to torsinAΔE protein than to torsinAwt. These findings provide functional evidence for the potential pathogenic nature of these rare sequence variants in the TOR1A gene, thus implicating these pathologies in the development of dystonia.
Collapse
Affiliation(s)
- Jasmin Hettich
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts; Department of Medical Genetics and Applied Genomics, University of Tuebingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Protein trafficking within the secretory pathway of mammalian cells is amenable to analysis by biochemical methods. This can be achieved by monitoring posttranslational modifications that occur naturally within the secretory pathway, or by measuring the delivery of cargo to the cell surface or extracellular medium. These approaches can be combined with additional manipulations such as specific temperature blocks that permit analysis of distinct trafficking steps. Biochemical analysis is advantageous in that it permits both a sensitive and quantitative measure of trafficking along the pathway. The methods discussed in this chapter permit the analysis of trafficking of both endogenous cargo proteins and ectopically expressed model cargos, which can be followed using either Western blotting or metabolic pulse-chase approaches. These methods are relatively straightforward and suitable for use in most modern cell biology laboratories. In addition to the well-established methods that we describe here in detail, we also refer to the development of more recent tailored approaches that add further to the arsenal of tools that can be used to assess trafficking in the secretory pathway.
Collapse
Affiliation(s)
- Peristera Roboti
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | | |
Collapse
|
45
|
Henderson MJ, Wires ES, Trychta KA, Richie CT, Harvey BK. SERCaMP: a carboxy-terminal protein modification that enables monitoring of ER calcium homeostasis. Mol Biol Cell 2014; 25:2828-39. [PMID: 25031430 PMCID: PMC4161517 DOI: 10.1091/mbc.e14-06-1141] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Depletion of ER calcium can lead to cell death and is implicated in numerous diseases. Secreted ER calcium-monitoring proteins (SERCaMPs) are secreted in response to ER calcium depletion. SERCaMPs identified ER calcium depletion in primary neurons exposed to glutamate, hyperthermia, and coxibs and in rat liver after a single exposure to thapsigargin. Endoplasmic reticulum (ER) calcium homeostasis is disrupted in diverse pathologies, including neurodegeneration, cardiovascular diseases, and diabetes. Temporally defining calcium dysregulation during disease progression, however, has been challenging. Here we describe secreted ER calcium-monitoring proteins (SERCaMPs), which allow for longitudinal monitoring of ER calcium homeostasis. We identified a carboxy-terminal modification that is sufficient to confer release of a protein specifically in response to ER calcium depletion. A Gaussia luciferase (GLuc)–based SERCaMP provides a simple and sensitive method to monitor ER calcium homeostasis in vitro or in vivo by analyzing culture medium or blood. GLuc-SERCaMPs revealed ER calcium depletion in rat primary neurons exposed to various ER stressors. In vivo, ER calcium disruption in rat liver was monitored over several days by repeated sampling of blood. Our results suggest that SERCaMPs will have broad applications for the long-term monitoring of ER calcium homeostasis and the development of therapeutic approaches to counteract ER calcium dysregulation.
Collapse
Affiliation(s)
- Mark J Henderson
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Emily S Wires
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Kathleen A Trychta
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Christopher T Richie
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| |
Collapse
|
46
|
Luft C, Freeman J, Elliott D, Al-Tamimi N, Kriston-Vizi J, Heintze J, Lindenschmidt I, Seed B, Ketteler R. Application of Gaussia luciferase in bicistronic and non-conventional secretion reporter constructs. BMC BIOCHEMISTRY 2014; 15:14. [PMID: 25007711 PMCID: PMC4099409 DOI: 10.1186/1471-2091-15-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 07/05/2014] [Indexed: 11/28/2022]
Abstract
Background Secreted luciferases are highly useful bioluminescent reporters for cell-based assays and drug discovery. A variety of secreted luciferases from marine organisms have been described that harbor an N-terminal signal peptide for release along the classical secretory pathway. Here, we have characterized the secretion of Gaussia luciferase in more detail. Results We describe three basic mechanisms by which GLUC can be released from cells: first, classical secretion by virtue of the N-terminal signal peptide; second, internal signal peptide-mediated secretion and third, non-conventional secretion in the absence of an N-terminal signal peptide. Non-conventional release of dNGLUC is not stress-induced, does not require autophagy and can be enhanced by growth factor stimulation. Furthermore, we have identified the golgi-associated, gamma adaptin ear containing, ARF binding protein 1 (GGA1) as a suppressor of release of dNGLUC. Conclusions Due to its secretion via multiple secretion pathways GLUC can find multiple applications as a research tool to study classical and non-conventional secretion. As GLUC can also be released from a reporter construct by internal signal peptide-mediated secretion it can be incorporated in a novel bicistronic secretion system.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Robin Ketteler
- Medical Research Council, Laboratory for Moleclar and Cell Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
47
|
Ashiru O, Howe JD, Butters TD. Nitazoxanide, an antiviral thiazolide, depletes ATP-sensitive intracellular Ca(2+) stores. Virology 2014; 462-463:135-48. [PMID: 24971706 DOI: 10.1016/j.virol.2014.05.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/01/2014] [Accepted: 05/14/2014] [Indexed: 12/31/2022]
Abstract
Nitazoxanide (NTZ) inhibits influenza, Japanese encephalitis, hepatitis B and hepatitis C virus replication but effects on the replication of other members of the Flaviviridae family has yet to be defined. The pestivirus bovine viral diarrhoea virus (BVDV) is a surrogate model for HCV infection and NTZ induced PKR and eIF2α phosphorylation in both uninfected and BVDV-infected cells. This led to the observation that NTZ depletes ATP-sensitive intracellular Ca(2+) stores. In addition to PKR and eIF2α phosphorylation, consequences of NTZ-mediated Ca(2+) mobilisation included induction of chronic sub-lethal ER stress as well as perturbation of viral protein N-linked glycosylation and trafficking. To adapt to NTZ-mediated ER stress, NTZ treated cells upregulated translation of Ca(2+)-binding proteins, including the ER chaperone Bip and the cytosolic pro-survival and anti-viral protein TCTP. Depletion of intracellular Ca(2+) stores is the primary consequence of NTZ treatment and is likely to underpin all antiviral mechanisms attributed to the thiazolide.
Collapse
Affiliation(s)
- Omodele Ashiru
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, Oxfordshire OX1 3QU, UK.
| | - Jonathon D Howe
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, Oxfordshire OX1 3QU, UK.
| | - Terry D Butters
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, Oxfordshire OX1 3QU, UK.
| |
Collapse
|
48
|
Nie J, Huang W, Wu X, Wang Y. Optimization and validation of a high throughput method for detecting neutralizing antibodies against human papillomavirus (HPV) based on pseudovirons. J Med Virol 2014; 86:1542-55. [DOI: 10.1002/jmv.23995] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2014] [Indexed: 01/28/2023]
Affiliation(s)
- Jianhui Nie
- College of Life Science; Jilin University; Changchun China
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of HIV/AIDS and Sex-Transmitted Virus Vaccines; National Institutes for Food and Drug Control (NIFDC); Beijing China
| | - Weijin Huang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of HIV/AIDS and Sex-Transmitted Virus Vaccines; National Institutes for Food and Drug Control (NIFDC); Beijing China
| | - Xueling Wu
- Key Laboratory of Ministry of Health for Research on Quality and Standardization of Biotech Products, Cell Collection and Research Center; National Institutes for Food and Drug Control (NIFDC); Beijing China
| | - Youchun Wang
- College of Life Science; Jilin University; Changchun China
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of HIV/AIDS and Sex-Transmitted Virus Vaccines; National Institutes for Food and Drug Control (NIFDC); Beijing China
| |
Collapse
|
49
|
miR-200a-mediated suppression of non-muscle heavy chain IIb inhibits meningioma cell migration and tumor growth in vivo. Oncogene 2014; 34:1790-8. [PMID: 24858044 DOI: 10.1038/onc.2014.120] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 04/13/2014] [Accepted: 04/14/2014] [Indexed: 12/14/2022]
Abstract
miR-200a has been implicated in the pathogenesis of meningiomas, one of the most common central nervous system tumors in humans. To identify how miR-200a contributes to meningioma pathogenesis at the molecular level, we used a comparative protein profiling approach using Gel-nanoLC-MS/MS and identified approximately 130 dysregulated proteins in miR-200a-overexpressing meningioma cells. Following the bioinformatic analysis to identify potential genes targeted by miR-200a, we focused on the non-muscle heavy chain IIb (NMHCIIb), and showed that miR-200a directly targeted NMHCIIb. Considering the key roles of NMHCIIb in cell division and cell migration, we aimed to identify whether miR-200a regulated these processes through NMHCIIb. We found that NMHCIIb overexpression partially rescued miR-200a-mediated inhibition of cell migration, as well as cell growth in vitro and in vivo. Moreover, siRNA-mediated silencing of NMHCIIb expression resulted in a similar migration phenotype in these cells and inhibited meningioma tumor growth in mice. Taken together, these results suggest that NMHCIIb might serve as a novel therapeutic target in meningiomas.
Collapse
|
50
|
Chen S, Xuan J, Couch L, Iyer A, Wu Y, Li QZ, Guo L. Sertraline induces endoplasmic reticulum stress in hepatic cells. Toxicology 2014; 322:78-88. [PMID: 24865413 DOI: 10.1016/j.tox.2014.05.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/08/2014] [Accepted: 05/16/2014] [Indexed: 10/25/2022]
Abstract
Sertraline is used for the treatment of depression, and is also used for the treatment of panic, obsessive-compulsive, and post-traumatic stress disorders. Previously, we have demonstrated that sertraline caused hepatic cytotoxicity, with mitochondrial dysfunction and apoptosis being underlying mechanisms. In this study, we used microarray and other biochemical and molecular analyses to identify endoplasmic reticulum (ER) stress as a novel molecular mechanism. HepG2 cells were exposed to sertraline and subjected to whole genome gene expression microarray analysis. Pathway analysis revealed that ER stress is among the significantly affected biological changes. We confirmed the increased expression of ER stress makers by real-time PCR and Western blots. The expression of typical ER stress markers such as PERK, IRE1α, and CHOP was significantly increased. To study better ER stress-mediated drug-induced liver toxicity; we established in vitro systems for monitoring ER stress quantitatively and efficiently, using Gaussia luciferase (Gluc) and secreted alkaline phosphatase (SEAP) as ER stress reporters. These in vitro systems were validated using well-known ER stress inducers. In these two reporter assays, sertraline inhibited the secretion of Gluc and SEAP. Moreover, we demonstrated that sertraline-induced apoptosis was coupled to ER stress and that the apoptotic effect was attenuated by 4-phenylbutyrate, a potent ER stress inhibitor. In addition, we showed that the MAP4K4-JNK signaling pathway contributed to the process of sertraline-induced ER stress. In summary, we demonstrated that ER stress is a mechanism of sertraline-induced liver toxicity.
Collapse
Affiliation(s)
- Si Chen
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR 72079, USA
| | - Jiekun Xuan
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR 72079, USA
| | - Letha Couch
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR 72079, USA
| | - Advait Iyer
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR 72079, USA; Biological Sciences, University of Maryland, Baltimore, MD 21250, USA
| | - Yuanfeng Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR 72079, USA
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR 72079, USA.
| |
Collapse
|