1
|
Jo A, Jung M, Mun JY, Kim YJ, Yoo JY. Membrane-tethered SCOTIN condensates elicit an endoplasmic reticulum stress response by sequestering luminal BiP. Cell Rep 2025; 44:115297. [PMID: 39946235 DOI: 10.1016/j.celrep.2025.115297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/28/2025] Open
Abstract
The endoplasmic reticulum (ER) stress response controls the balance between cellular survival and death. Here, we implicate SCOTIN, an interferon-inducible ER protein, in activating the ER stress response and modulating cell fate through its proline-rich domain (PRD)-mediated cytosolic condensation. SCOTIN overexpression leads to the formation of condensates enveloping multiple layers of the ER, accompanied by morphological signs of organelle stress. Luminal BiP chaperone proteins are sequestered within these SCOTIN condensates, which elicit ER stress responses. The colocalization of luminal BiP with SCOTIN is strictly contingent upon the PRD-mediated condensation of SCOTIN in the cytosolic compartment, closely associated with the ER membrane. The cysteine-rich domain (CRD) of SCOTIN, along with the condensation-prone PRD domain, is required for ER stress induction. We propose that membrane-associated condensation transduces signals across the ER membrane, leading to the induction of BiP assembly and the ER stress response.
Collapse
Affiliation(s)
- Areum Jo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Minkyo Jung
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Ji Young Mun
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Young Jin Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Joo-Yeon Yoo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
2
|
Chadwick SR, Stack-Couture S, Berg MD, Di Gregorio S, Lung B, Genereaux J, Moir RD, Brandl CJ, Willis IM, Snapp EL, Lajoie P. TUDCA modulates drug bioavailability to regulate resistance to acute ER stress in Saccharomyces cerevisiae. Mol Biol Cell 2025; 36:ar13. [PMID: 39661468 PMCID: PMC11809307 DOI: 10.1091/mbc.e24-04-0147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024] Open
Abstract
Cells counter accumulation of misfolded secretory proteins in the endoplasmic reticulum (ER) through activation of the Unfolded Protein Response (UPR). Small molecules termed chemical chaperones can promote protein folding to alleviate ER stress. The bile acid tauroursodeoxycholic acid (TUDCA) has been described as a chemical chaperone. While promising in models of protein folding diseases, TUDCA's mechanism of action remains unclear. Here, we found TUDCA can rescue growth of yeast treated with the ER stressor tunicamycin (Tm), even in the absence of a functional UPR. In contrast, TUDCA failed to rescue growth on other ER stressors. Nor could TUDCA attenuate chronic UPR associated with specific gene deletions or overexpression of a misfolded mutant secretory protein. Neither pretreatment with nor delayed addition of TUDCA conferred protection against Tm. Importantly, attenuation of Tm-induced toxicity required TUDCA's critical micelle forming concentration, suggesting a mechanism where TUDCA directly sequesters drugs. Indeed, in several assays, TUDCA-treated cells closely resembled cells treated with lower doses of Tm. In addition, we found TUDCA can inhibit dyes from labeling intracellular compartments. Thus, our study challenges the model of TUDCA as a chemical chaperone and suggests that TUDCA decreases drug bioavailability, allowing cells to adapt to ER stress.
Collapse
Affiliation(s)
- Sarah R. Chadwick
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Samuel Stack-Couture
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Matthew D. Berg
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Sonja Di Gregorio
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Bryan Lung
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Julie Genereaux
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Robyn D. Moir
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Christopher J. Brandl
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Ian M. Willis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Erik L. Snapp
- Janelia Research Campus of the Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Patrick Lajoie
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
- Children's Health Research Institute, Lawson Health Research Institute, London, Ontario N6C 2V5, Canada
| |
Collapse
|
3
|
Fakim A, Maatouk BI, Maiti B, Dey A, Alotaiby SH, Moosa BA, Lin W, Khashab NM. Flaring Inflammation and ER Stress by an Organelle-Specific Fluorescent Cage. Adv Healthc Mater 2024; 13:e2401117. [PMID: 38848965 DOI: 10.1002/adhm.202401117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/21/2024] [Indexed: 06/09/2024]
Abstract
The endoplasmic reticulum (ER) plays an important role in protein synthesis and its disruption can cause protein unfolding and misfolding. Accumulation of such proteins leads to ER stress, which ultimately promotes many diseases. Routine screening of ER activity in immune cells can flag serious conditions at early stages, but the current clinically used bio-probes have limitations. Herein, an ER-specific fluorophore based on a biocompatible benzothiadiazole-imine cage (BTD-cage) with excellent photophysical properties is developed. The cage outperforms commercially available ER stains in long-term live cell imaging with no fading or photobleaching over time. The cage is responsive to different levels of ER stress where its fluorescence increases accordingly. Incorporating the bio-probe into an immune disorder model, a 6-, 21-, and 48-fold increase in intensity is shown in THP-1, Raw 246.7, and Jurkat cells, respectively (within 15 min). These results strongly support that this system can be used for rapid visual and selective detection of ER stress. It is envisaged that tailoring molecular interactions and molecular recognition using supramolecular improved fluorophores can expand the library of biological probes for enhanced selectivity and targetability toward cellular organelles.
Collapse
Affiliation(s)
- Aliyah Fakim
- Chemistry Program, Physical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Batoul I Maatouk
- Chemistry Program, Physical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Bappa Maiti
- Chemistry Program, Physical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Avishek Dey
- Chemistry Program, Physical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Shahad H Alotaiby
- Chemistry Program, Physical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Basem A Moosa
- Chemistry Program, Physical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Weibin Lin
- Chemistry Program, Physical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Niveen M Khashab
- Chemistry Program, Physical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| |
Collapse
|
4
|
Sacco M, Testa MF, Ferretti A, Basso M, Lancellotti S, Tardugno M, Di Gennaro L, Concolino P, Minucci A, Spoliti C, Branchini A, De Cristofaro R. An integrated multitool analysis contributes elements to interpreting unclassified factor IX missense variants associated with hemophilia B. J Thromb Haemost 2024; 22:2724-2738. [PMID: 39019441 DOI: 10.1016/j.jtha.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/10/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Dissection of genotype-phenotype relationships in hemophilia B (HB) is particularly relevant for challenging (mild HB) or for HB-associated but unclassified factor (F)IX missense variants. OBJECTIVE To contribute elements to interpret unclassified HB-associated FIX missense variants by a multiple-level approach upon identification of a reported, but uncharacterized, FIX missense variant associated with mild HB. METHODS Molecular modeling of wild-type and V92A FIX variants, expression studies in HEK293 cells with evaluation of protein (ELISA, western blotting) and activity (activated partial thromboplastin time-based/chromogenic assays) levels after recombinant expression, and multiple prediction tools. RESULTS The F9(NM_000133.4):c.275T>C (p.V92A) variant was found in a mild HB patient (antigen, 45.4 U/dL; coagulant activity, 23.6 IU/dL; specific activity, 0.52). Newly generated molecular models showed alterations in Gla/EGF1-EGF2 domain conformation impacting Ca++ affinity and protein-protein interactions with activated factor XI (FXIa). Multitool analysis indicated a moderate impact on protein structure/function of the valine-to-alanine substitution, in accordance with patient and modeling data. Expression studies on the V92A variant showed a specific activity (0.49 ± 0.07; wild-type, 1.0 ± 0.1) recapitulating that of the natural variant, and pointed toward a moderate activation impairment as the main determinant underlying the p.V92A defect. The validated multitool approach, integrated with evidence-based data, was challenged on a panel (n = 9) of unclassified FIX missense variants, which resulted in inferred protein (secretion/function) outputs and HB severity. CONCLUSION The rational integration of multitool and multiparameter analyses contributed elements to interpret genotype/phenotype relationships of unclassified FIX missense variants, with implications for diagnosis, management, and treatment of HB patients, and potentially translatable into other human disorders.
Collapse
Affiliation(s)
- Monica Sacco
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
| | - Maria Francesca Testa
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Antonietta Ferretti
- Center for Hemorrhagic and Thrombotic Diseases, Foundation University Hospital "A. Gemelli" IRCCS, Rome, Italy
| | - Maria Basso
- Center for Hemorrhagic and Thrombotic Diseases, Foundation University Hospital "A. Gemelli" IRCCS, Rome, Italy
| | - Stefano Lancellotti
- Center for Hemorrhagic and Thrombotic Diseases, Foundation University Hospital "A. Gemelli" IRCCS, Rome, Italy
| | - Maira Tardugno
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
| | - Leonardo Di Gennaro
- Center for Hemorrhagic and Thrombotic Diseases, Foundation University Hospital "A. Gemelli" IRCCS, Rome, Italy
| | - Paola Concolino
- Molecular and Genomic Diagnostics Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Angelo Minucci
- Molecular and Genomic Diagnostics Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Claudia Spoliti
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
| | - Alessio Branchini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.
| | - Raimondo De Cristofaro
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy; Center for Hemorrhagic and Thrombotic Diseases, Foundation University Hospital "A. Gemelli" IRCCS, Rome, Italy.
| |
Collapse
|
5
|
Najarro G, Brackett K, Woosley H, Dorman LC, Turon-Lagot V, Khadka S, Faeldonea C, Moreno OK, Negron AR, Love C, Ward R, Langelier C, McCarthy F, Gonzalez C, Elias JE, Gardner BM, Arias C. BiP/GRP78 is a pro-viral factor for diverse dsDNA viruses that promotes the survival and proliferation of cells upon KSHV infection. PLoS Pathog 2024; 20:e1012660. [PMID: 39471213 PMCID: PMC11548844 DOI: 10.1371/journal.ppat.1012660] [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: 07/26/2024] [Revised: 11/08/2024] [Accepted: 10/11/2024] [Indexed: 11/01/2024] Open
Abstract
The Endoplasmic Reticulum (ER)-resident HSP70 chaperone BiP (HSPA5) plays a crucial role in maintaining and restoring protein folding homeostasis in the ER. BiP's function is often dysregulated in cancer and virus-infected cells, conferring pro-oncogenic and pro-viral advantages. We explored BiP's functions during infection by the Kaposi's sarcoma-associated herpesvirus (KSHV), an oncogenic gamma-herpesvirus associated with cancers of immunocompromised patients. Our findings reveal that BiP protein levels are upregulated in infected epithelial cells during the lytic phase of KSHV infection. This upregulation occurs independently of the unfolded protein response (UPR), a major signaling pathway that regulates BiP availability. Genetic and pharmacological inhibition of BiP halts KSHV viral replication and reduces the proliferation and survival of KSHV-infected cells. Notably, inhibition of BiP limits the spread of other alpha- and beta-herpesviruses and poxviruses with minimal toxicity for normal cells. Our work suggests that BiP is a potential target for developing broad-spectrum antiviral therapies against double-stranded DNA viruses and a promising candidate for therapeutic intervention in KSHV-related malignancies.
Collapse
Affiliation(s)
- Guillermo Najarro
- University of California, Santa Barbara, California, United States of America
| | - Kevin Brackett
- University of California, Santa Barbara, California, United States of America
| | - Hunter Woosley
- Chan Zuckerberg BioHub, San Francisco, California, United States of America
| | - Leah C. Dorman
- Chan Zuckerberg BioHub, San Francisco, California, United States of America
| | | | - Sudip Khadka
- Chan Zuckerberg BioHub, San Francisco, California, United States of America
| | - Catya Faeldonea
- University of California, Santa Barbara, California, United States of America
| | | | | | - Christina Love
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Ryan Ward
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Charles Langelier
- Chan Zuckerberg BioHub, San Francisco, California, United States of America
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Frank McCarthy
- Chan Zuckerberg BioHub, San Francisco, California, United States of America
| | - Carlos Gonzalez
- Chan Zuckerberg BioHub, San Francisco, California, United States of America
| | - Joshua E. Elias
- Chan Zuckerberg BioHub, San Francisco, California, United States of America
| | - Brooke M. Gardner
- University of California, Santa Barbara, California, United States of America
| | - Carolina Arias
- University of California, Santa Barbara, California, United States of America
- Chan Zuckerberg BioHub, San Francisco, California, United States of America
| |
Collapse
|
6
|
Besse A, Sedlarikova L, Buechler L, Kraus M, Yang CH, Strakova N, Soucek K, Navratil J, Svoboda M, Welm AL, Joerger M, Driessen C, Besse L. HIV-protease inhibitors potentiate the activity of carfilzomib in triple-negative breast cancer. Br J Cancer 2024; 131:918-930. [PMID: 38969867 PMCID: PMC11368961 DOI: 10.1038/s41416-024-02774-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 06/08/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Resistance to chemotherapy is a major problem in the treatment of patients with triple-negative breast cancer (TNBC). Preclinical data suggest that TNBC is dependent on proteasomes; however, clinical observations indicate that the efficacy of proteasome inhibitors in TNBC may be limited, suggesting the need for combination therapies. METHODS We compared bortezomib and carfilzomib and their combinations with nelfinavir and lopinavir in TNBC cell lines and primary cells with regard to their cytotoxic activity, functional proteasome inhibition, and induction of the unfolded protein response (UPR). Furthermore, we evaluated the involvement of sXBP1, ABCB1, and ABCG2 in the cytotoxic activity of drug combinations. RESULTS Carfilzomib, via proteasome β5 + β2 inhibition, is more cytotoxic in TNBC than bortezomib, which inhibits β5 + β1 proteasome subunits. The cytotoxicity of carfilzomib was significantly potentiated by nelfinavir or lopinavir. Carfilzomib with lopinavir induced endoplasmic reticulum stress and pro-apoptotic UPR through the accumulation of excess proteasomal substrate protein in TNBC in vitro. Moreover, lopinavir increased the intracellular availability of carfilzomib by inhibiting carfilzomib export from cells that express high levels and activity of ABCB1, but not ABCG2. CONCLUSION Proteasome inhibition by carfilzomib combined with nelfinavir/lopinavir represents a potential treatment option for TNBC, warranting further investigation.
Collapse
Affiliation(s)
- Andrej Besse
- Laboratory of Experimental Oncology, Department of Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, 9000, Switzerland
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, 62500, Czech Republic
| | - Lenka Sedlarikova
- Babak Myeloma Group, Department of Pathological Physiology, Masaryk University, Brno, 62500, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, 62500, Czech Republic
| | - Lorina Buechler
- Laboratory of Experimental Oncology, Department of Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, 9000, Switzerland
| | - Marianne Kraus
- Laboratory of Experimental Oncology, Department of Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, 9000, Switzerland
| | - Chieh-Hsiang Yang
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicol Strakova
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, 612 00, Czech Republic
- Veterinary Research Institute, Brno, 62500, Czech Republic
| | - Karel Soucek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, 612 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Jiri Navratil
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, 62500, Czech Republic
- Faculty of Medicine, Masaryk University, Brno, 62500, Czech Republic
| | - Marek Svoboda
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, 62500, Czech Republic
- Faculty of Medicine, Masaryk University, Brno, 62500, Czech Republic
| | - Alana L Welm
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Markus Joerger
- Department of Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, 9000, Switzerland
| | - Christoph Driessen
- Laboratory of Experimental Oncology, Department of Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, 9000, Switzerland
- Department of Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, 9000, Switzerland
| | - Lenka Besse
- Laboratory of Experimental Oncology, Department of Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, 9000, Switzerland.
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, 62500, Czech Republic.
| |
Collapse
|
7
|
Hendershot LM, Buck TM, Brodsky JL. The Essential Functions of Molecular Chaperones and Folding Enzymes in Maintaining Endoplasmic Reticulum Homeostasis. J Mol Biol 2024; 436:168418. [PMID: 38143019 PMCID: PMC12015986 DOI: 10.1016/j.jmb.2023.168418] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
It has been estimated that up to one-third of the proteins encoded by the human genome enter the endoplasmic reticulum (ER) as extended polypeptide chains where they undergo covalent modifications, fold into their native structures, and assemble into oligomeric protein complexes. The fidelity of these processes is critical to support organellar, cellular, and organismal health, and is perhaps best underscored by the growing number of disease-causing mutations that reduce the fidelity of protein biogenesis in the ER. To meet demands encountered by the diverse protein clientele that mature in the ER, this organelle is populated with a cadre of molecular chaperones that prevent protein aggregation, facilitate protein disulfide isomerization, and lower the activation energy barrier of cis-trans prolyl isomerization. Components of the lectin (glycan-binding) chaperone system also reside within the ER and play numerous roles during protein biogenesis. In addition, the ER houses multiple homologs of select chaperones that can recognize and act upon diverse peptide signatures. Moreover, redundancy helps ensure that folding-compromised substrates are unable to overwhelm essential ER-resident chaperones and enzymes. In contrast, the ER in higher eukaryotic cells possesses a single member of the Hsp70, Hsp90, and Hsp110 chaperone families, even though several homologs of these molecules reside in the cytoplasm. In this review, we discuss specific functions of the many factors that maintain ER quality control, highlight some of their interactions, and describe the vulnerabilities that arise from the absence of multiple members of some chaperone families.
Collapse
Affiliation(s)
- Linda M Hendershot
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States.
| | - Teresa M Buck
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, United States
| |
Collapse
|
8
|
Pontisso I, Ornelas-Guevara R, Chevet E, Combettes L, Dupont G. Gradual ER calcium depletion induces a progressive and reversible UPR signaling. PNAS NEXUS 2024; 3:pgae229. [PMID: 38933930 PMCID: PMC11200134 DOI: 10.1093/pnasnexus/pgae229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 05/29/2024] [Indexed: 06/28/2024]
Abstract
The unfolded protein response (UPR) is a widespread signal transduction pathway triggered by endoplasmic reticulum (ER) stress. Because calcium (Ca2+) is a key factor in the maintenance of ER homeostasis, massive Ca2+ depletion of the ER is a potent inducer of ER stress. Although moderate changes in ER Ca2+ drive the ubiquitous Ca2+ signaling pathways, a possible incremental relationship between UPR activation and Ca2+ changes has yet to be described. Here, we determine the sensitivity and time-dependency of activation of the three ER stress sensors, inositol-requiring protein 1 alpha (IRE1α), protein kinase R-like ER kinase (PERK), and activating transcription factor 6 alpha (ATF6α) in response to controlled changes in the concentration of ER Ca2+ in human cultured cells. Combining Ca2+ imaging, fluorescence recovery after photobleaching experiments, biochemical analyses, and mathematical modeling, we uncover a nonlinear rate of activation of the IRE1α branch of UPR, as compared to the PERK and ATF6α branches that become activated gradually with time and are sensitive to more important ER Ca2+ depletions. However, the three arms are all activated within a 1 h timescale. The model predicted the deactivation of PERK and IRE1α upon refilling the ER with Ca2+. Accordingly, we showed that ER Ca2+ replenishment leads to the complete reversion of IRE1α and PERK phosphorylation in less than 15 min, thus revealing the highly plastic character of the activation of the upstream UPR sensors. In conclusion, our results reveal a dynamic and dose-sensitive Ca2+-dependent activation/deactivation cycle of UPR induction, which could tightly control cell fate upon acute and/or chronic stress.
Collapse
Affiliation(s)
- Ilaria Pontisso
- U1282 “Calcium Signaling and Microbial Infections”, Institut de Biologie Intégrative de la Cellule (I2BC)—Université Paris-Saclay, Gif-Sur-Yvette 91190, France
| | - Roberto Ornelas-Guevara
- Unit of Theoretical Chronobiology, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - Eric Chevet
- Inserm U1242 Université de Rennes, 35000 Rennes, France
- Centre de Lutte Contre le Cancer Eugène Marquis, 35042 Rennes, France
| | - Laurent Combettes
- U1282 “Calcium Signaling and Microbial Infections”, Institut de Biologie Intégrative de la Cellule (I2BC)—Université Paris-Saclay, Gif-Sur-Yvette 91190, France
| | - Geneviève Dupont
- Unit of Theoretical Chronobiology, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
| |
Collapse
|
9
|
Liu Z, Liu G, Ha DP, Wang J, Xiong M, Lee AS. ER chaperone GRP78/BiP translocates to the nucleus under stress and acts as a transcriptional regulator. Proc Natl Acad Sci U S A 2023; 120:e2303448120. [PMID: 37487081 PMCID: PMC10400976 DOI: 10.1073/pnas.2303448120] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/08/2023] [Indexed: 07/26/2023] Open
Abstract
Cancer cells are commonly subjected to endoplasmic reticulum (ER) stress. To gain survival advantage, cancer cells exploit the adaptive aspects of the unfolded protein response such as upregulation of the ER luminal chaperone GRP78. The finding that when overexpressed, GRP78 can escape to other cellular compartments to gain new functions regulating homeostasis and tumorigenesis represents a paradigm shift. Here, toward deciphering the mechanisms whereby GRP78 knockdown suppresses EGFR transcription, we find that nuclear GRP78 is prominent in cancer and stressed cells and uncover a nuclear localization signal critical for its translocation and nuclear activity. Furthermore, nuclear GRP78 can regulate expression of genes and pathways, notably those important for cell migration and invasion, by interacting with and inhibiting the activity of the transcriptional repressor ID2. Our study reveals a mechanism for cancer cells to respond to ER stress via transcriptional regulation mediated by nuclear GRP78 to adopt an invasive phenotype.
Collapse
Affiliation(s)
- Ze Liu
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA90033
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Guanlin Liu
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA90033
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Dat P. Ha
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA90033
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Justin Wang
- Department of Molecular Medicine, Scripps Research, La Jolla, CA92037
| | - Min Xiong
- Department of System Biology, Beckman Research Institute, City of Hope, Duarte, CA91010
| | - Amy S. Lee
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA90033
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| |
Collapse
|
10
|
Kotler JLM, Street TO. Mechanisms of Protein Quality Control in the Endoplasmic Reticulum by a Coordinated Hsp40-Hsp70-Hsp90 System. Annu Rev Biophys 2023; 52:509-524. [PMID: 37159299 DOI: 10.1146/annurev-biophys-111622-091309] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The Hsp40, Hsp70, and Hsp90 chaperone families are ancient, highly conserved, and critical to cellular protein homeostasis. Hsp40 chaperones can transfer their protein clients to Hsp70, and Hsp70 can transfer clients to Hsp90, but the functional benefits of these transfers are unclear. Recent structural and mechanistic work has opened up the possibility of uncovering how Hsp40, Hsp70, and Hsp90 work together as unified system. In this review, we compile mechanistic data on the ER J-domain protein 3 (ERdj3) (an Hsp40), BiP (an Hsp70), and Grp94 (an Hsp90) chaperones within the endoplasmic reticulum; what is known about how these chaperones work together; and gaps in this understanding. Using calculations, we examine how client transfer could impact the solubilization of aggregates, the folding of soluble proteins, and the triage decisions by which proteins are targeted for degradation. The proposed roles of client transfer among Hsp40-Hsp70-Hsp90 chaperones are new hypotheses, and we discuss potential experimental tests of these ideas.
Collapse
Affiliation(s)
- Judy L M Kotler
- Department of Biochemistry, Brandeis University, Waltham, Massachusetts, USA;
| | - Timothy O Street
- Department of Biochemistry, Brandeis University, Waltham, Massachusetts, USA;
| |
Collapse
|
11
|
Fang H, Hu L, Chen Q, Geng S, Qiu K, Wang C, Hao M, Tian Z, Chen H, Liu L, Guan JL, Chen Y, Dong L, Guo Z, He W, Diao J. An ER-targeted "reserve-release" fluorogen for topological quantification of reticulophagy. Biomaterials 2023; 292:121929. [PMID: 36455487 DOI: 10.1016/j.biomaterials.2022.121929] [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: 03/27/2022] [Revised: 11/09/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022]
Abstract
The endoplasmic reticulum's (ER) dynamic nature, essential for maintaining cellular homeostasis, can be influenced by stress-induced damage, which can be assessed by examining the morphology of ER dynamics and, more locally, ER properties such as hydrophobicity, viscosity, and polarity. Although numerous ER-specific chemical probes have been developed to monitor the ER's physical and chemical parameters, the quantitative detection and super-resolution imaging of its local hydrophobicity have yet to be explored. Here, we describe a photostable ER-targeted probe with high signal-to-noise ratio for super-resolution imaging that can specifically respond to changes in ER hydrophobicity under stress based on a "reserve-release" mechanism. The probe shows an excellent ability to target ER over commercial ER dyes and can be used to track local changes of hydrophobicity by fluorescence intensity and morphology during the selective autophagy of ER (i.e., reticulophagy). By correlating the level and location of ER damage with the distribution of fluorescence intensity, we were able to assess reticulophagy at the subcellular level. Beyond that, we developed a topological analytical tool adaptable to any ER probe for detecting structural changes in ER and thus quantitatively identifying reticulophagy. The algorithm-assisted tool can also be adapted to a wide range of molecular probes and organelles. Altogether, the new probe and analytical strategy described here show promise for the quantitative detection and analysis of subtle ER damage and stress.
Collapse
Affiliation(s)
- Hongbao Fang
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Lianting Hu
- Medical Big Data Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangzhou, 510080, China; School of Information Management, Wuhan University, Wuhan 430072, China
| | - Qixin Chen
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Shanshan Geng
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Kangqiang Qiu
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Chengjun Wang
- Sinopec Shengli Petroleum Engineering Limited Company, Dongying, 257000, China
| | - Mingang Hao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Zhiqi Tian
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Huimin Chen
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Lei Liu
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Yuncong Chen
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Lei Dong
- School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Weijiang He
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
12
|
Walter F, D’Orsi B, Jagannathan A, Dussmann H, Prehn JHM. BOK controls ER proteostasis and physiological ER stress responses in neurons. Front Cell Dev Biol 2022; 10:915065. [PMID: 36060797 PMCID: PMC9434404 DOI: 10.3389/fcell.2022.915065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
The Bcl-2 family proteins BAK and BAX control the crucial step of pore formation in the mitochondrial outer membrane during intrinsic apoptosis. Bcl-2-related ovarian killer (BOK) is a Bcl-2 family protein with a high sequence similarity to BAK and BAX. However, intrinsic apoptosis can proceed in the absence of BOK. Unlike BAK and BAX, BOK is primarily located on the endoplasmic reticulum (ER) and Golgi membranes, suggesting a role for BOK in regulating ER homeostasis. In this study, we report that BOK is required for a full ER stress response. Employing previously characterized fluorescent protein-based ER stress reporter cell systems, we show that BOK-deficient cells have an attenuated response to ER stress in all three signaling branches of the unfolded protein response. Fluo-4-based confocal Ca2+ imaging revealed that disruption of ER proteostasis in BOK-deficient cells was not linked to altered ER Ca2+ levels. Fluorescence recovery after photobleaching (FRAP) experiments using GRP78/BiP-eGFP demonstrated that GRP78 motility was significantly lower in BOK-deficient cells. This implied that less intraluminal GRP78 was freely available and more of the ER chaperone bound to unfolded proteins. Collectively, these experiments suggest a new role for BOK in the protection of ER proteostasis and cellular responses to ER stress.
Collapse
Affiliation(s)
- Franziska Walter
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
- SFI FutureNeuro Research Centre, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Beatrice D’Orsi
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
- Institute of Neuroscience, Italian National Research Council, Pisa, Italy
| | - Anagha Jagannathan
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Jochen H. M. Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
- SFI FutureNeuro Research Centre, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
- *Correspondence: Jochen H. M. Prehn,
| |
Collapse
|
13
|
Xiong X, Huang KB, Wang Y, Cao B, Luo Y, Chen H, Yang Y, Long Y, Liu M, Chan ASC, Liang H, Zou T. Target Profiling of an Iridium(III)-Based Immunogenic Cell Death Inducer Unveils the Engagement of Unfolded Protein Response Regulator BiP. J Am Chem Soc 2022; 144:10407-10416. [PMID: 35658433 DOI: 10.1021/jacs.2c02435] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clinical chemotherapeutic drugs have occasionally been observed to induce antitumor immune responses beyond the direct cytotoxicity. Such effects are coined as immunogenic cell death (ICD), representing a "second hit" from the host immune system to tumor cells. Although chemo-immunotherapy is highly promising, ICD inducers remain sparse with vague drug-target mechanisms. Here, we report an endoplasmic reticulum stress-inducing cyclometalated Ir(III)-bisNHC complex (1a) as a new ICD inducer, and based on this compound, a clickable photoaffinity probe was designed for target identification, which unveiled the engagement of the master regulator protein BiP (binding immunoglobulin protein)/GRP78 of the unfolded protein response pathway. This has been confirmed by a series of cellular and biochemical studies including fluorescence microscopy, cellular thermal shift assay, enzymatic assays, and so forth, showing the capability of 1a for BiP destabilization. Notably, besides 1a, the previously reported ICD inducers including KP1339, mitoxantrone, and oxaliplatin were also found to engage BiP interaction, suggesting the important role of BiP in eliciting anticancer immunity. We believe that the ICD-related target information in this work will help to understand the mode of action of ICD that is beneficial to designing new ICD agents with high specificity and improved efficacy.
Collapse
Affiliation(s)
- Xiaolin Xiong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Ke-Bin Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmacy, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Yuan Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Bei Cao
- Warshel Institute for Computational Biology, and General Education Division, The Chinese University of Hong Kong, Shenzhen 518172, P. R. China
| | - Yunli Luo
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Huowen Chen
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Yan Yang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Yan Long
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Moyi Liu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Albert S C Chan
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmacy, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Taotao Zou
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| |
Collapse
|
14
|
Liu Y, Teng L, Yin B, Meng H, Yin X, Huan S, Song G, Zhang XB. Chemical Design of Activatable Photoacoustic Probes for Precise Biomedical Applications. Chem Rev 2022; 122:6850-6918. [PMID: 35234464 DOI: 10.1021/acs.chemrev.1c00875] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Photoacoustic (PA) imaging technology, a three-dimensional hybrid imaging modality that integrates the advantage of optical and acoustic imaging, has great application prospects in molecular imaging due to its high imaging depth and resolution. To endow PA imaging with the ability for real-time molecular visualization and precise biomedical diagnosis, numerous activatable molecular PA probes which can specifically alter their PA intensities upon reacting with the targets or biological events of interest have been developed. This review highlights the recent developments of activatable PA probes for precise biomedical applications including molecular detection of the biotargets and imaging of the biological events. First, the generation mechanism of PA signals will be given, followed by a brief introduction to contrast agents used for PA probe design. Then we will particularly summarize the general design principles for the alteration of PA signals and activatable strategies for developing precise PA probes. Furthermore, we will give a detailed discussion of activatable PA probes in molecular detection and biomedical imaging applications in living systems. At last, the current challenges and outlooks of future PA probes will be discussed. We hope that this review will stimulate new ideas to explore the potentials of activatable PA probes for precise biomedical applications in the future.
Collapse
Affiliation(s)
- Yongchao Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Lili Teng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Baoli Yin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Hongmin Meng
- College of Chemistry, Green Catalysis Center, Zhengzhou University, Zhengzhou 450001, China
| | - Xia Yin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Shuangyan Huan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
15
|
Navarro-Tapia E, Pérez-Torrado R. Indirect Methods To Measure Unfolded Proteins In Living Cells Using Fluorescent Proteins. Methods Mol Biol 2022; 2378:31-44. [PMID: 34985692 DOI: 10.1007/978-1-0716-1732-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In the study of the unfolded protein response pathway, it is essential to determine the amount of unfolded proteins that the cell is accumulating. Besides being essential it is one of the most challenging technique because of the difficulty to detect unfolded proteins without producing protein denaturation with the method itself. Thus, indirect methods became very useful as the use of fluorescent proteins. In this chapter, we present some of the most used methods to indirectly measure protein folding in living cells using fluorescent proteins.
Collapse
|
16
|
Amini M, Chang Y, Wissenbach U, Flockerzi V, Schlenstedt G, Beck A. Activity of the yeast vacuolar TRP channel TRPY1 is inhibited by Ca 2+-calmodulin binding. J Biol Chem 2021; 297:101126. [PMID: 34461097 PMCID: PMC8449268 DOI: 10.1016/j.jbc.2021.101126] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential (TRP) cation channels, which are conserved across mammals, flies, fish, sea squirts, worms, and fungi, essentially contribute to cellular Ca2+ signaling. The activity of the unique TRP channel in yeast, TRP yeast channel 1 (TRPY1), relies on the vacuolar and cytoplasmic Ca2+ concentration. However, the mechanism(s) of Ca2+-dependent regulation of TRPY1 and possible contribution(s) of Ca2+-binding proteins are yet not well understood. Our results demonstrate a Ca2+-dependent binding of yeast calmodulin (CaM) to TRPY1. TRPY1 activity was increased in the cmd1–6 yeast strain, carrying a non–Ca2+-binding CaM mutant, compared with the parent strain expressing wt CaM (Cmd1). Expression of Cmd1 in cmd1–6 yeast rescued the wt phenotype. In addition, in human embryonic kidney 293 cells, hypertonic shock-induced TRPY1-dependent Ca2+ influx and Ca2+ release were increased by the CaM antagonist ophiobolin A. We found that coexpression of mammalian CaM impeded the activity of TRPY1 by reinforcing effects of endogenous CaM. Finally, inhibition of TRPY1 by Ca2+–CaM required the cytoplasmic amino acid stretch E33–Y92. In summary, our results show that TRPY1 is under inhibitory control of Ca2+–CaM and that mammalian CaM can replace yeast CaM for this inhibition. These findings add TRPY1 to the innumerable cellular proteins, which include a variety of ion channels, that use CaM as a constitutive or dissociable Ca2+-sensing subunit, and contribute to a better understanding of the modulatory mechanisms of Ca2+–CaM.
Collapse
Affiliation(s)
- Mahnaz Amini
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Deutschland; Department of Medical Biochemistry and Molecular Biology/PZMS, Medical School, Saarland University, Homburg, Germany
| | - Yiming Chang
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Deutschland; Department of Medical Biochemistry and Molecular Biology/PZMS, Medical School, Saarland University, Homburg, Germany
| | - Ulrich Wissenbach
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Deutschland
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Deutschland
| | - Gabriel Schlenstedt
- Department of Medical Biochemistry and Molecular Biology/PZMS, Medical School, Saarland University, Homburg, Germany
| | - Andreas Beck
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Deutschland.
| |
Collapse
|
17
|
Comparative analysis of the coordinated motion of Hsp70s from different organelles observed by single-molecule three-color FRET. Proc Natl Acad Sci U S A 2021; 118:2025578118. [PMID: 34389669 DOI: 10.1073/pnas.2025578118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cellular function depends on the correct folding of proteins inside the cell. Heat-shock proteins 70 (Hsp70s), being among the first molecular chaperones binding to nascently translated proteins, aid in protein folding and transport. They undergo large, coordinated intra- and interdomain structural rearrangements mediated by allosteric interactions. Here, we applied a three-color single-molecule Förster resonance energy transfer (FRET) combined with three-color photon distribution analysis to compare the conformational cycle of the Hsp70 chaperones DnaK, Ssc1, and BiP. By capturing three distances simultaneously, we can identify coordinated structural changes during the functional cycle. Besides the known conformations of the Hsp70s with docked domains and open lid and undocked domains with closed lid, we observed additional intermediate conformations and distance broadening, suggesting flexibility of the Hsp70s in adopting the states in a coordinated fashion. Interestingly, the difference of this distance broadening varied between DnaK, Ssc1, and BiP. Study of their conformational cycle in the presence of substrate peptide and nucleotide exchange factors strengthened the observation of additional conformational intermediates, with BiP showing coordinated changes more clearly compared to DnaK and Ssc1. Additionally, DnaK and BiP were found to differ in their selectivity for nucleotide analogs, suggesting variability in the recognition mechanism of their nucleotide-binding domains for the different nucleotides. By using three-color FRET, we overcome the limitations of the usual single-distance approach in single-molecule FRET, allowing us to characterize the conformational space of proteins in higher detail.
Collapse
|
18
|
A virtuous cycle operated by ERp44 and ERGIC-53 guarantees proteostasis in the early secretory compartment. iScience 2021; 24:102244. [PMID: 33763635 PMCID: PMC7973864 DOI: 10.1016/j.isci.2021.102244] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/01/2021] [Accepted: 02/25/2021] [Indexed: 01/13/2023] Open
Abstract
The composition of the secretome depends on the combined action of cargo receptors that facilitate protein transport and sequential checkpoints that restrict it to native conformers. Acting after endoplasmic reticulum (ER)-resident chaperones, ERp44 retrieves its clients from downstream compartments. To guarantee efficient quality control, ERp44 should exit the ER as rapidly as its clients, or more. Here, we show that appending ERp44 to different cargo proteins increases their secretion rates. ERp44 binds the cargo receptor ER-Golgi intermediate compartment (ERGIC)-53 in the ER to negotiate preferential loading into COPII vesicles. Silencing ERGIC-53, or competing for its COPII binding with 4-phenylbutyrate, causes secretion of Prdx4, an enzyme that relies on ERp44 for intracellular localization. In more acidic, zinc-rich downstream compartments, ERGIC-53 releases its clients and ERp44, which can bind and retrieve non-native conformers via KDEL receptors. By coupling the transport of cargoes and inspector proteins, cells ensure efficiency and fidelity of secretion.
Collapse
|
19
|
Gomez-Navarro N, Melero A, Li XH, Boulanger J, Kukulski W, Miller EA. Cargo crowding contributes to sorting stringency in COPII vesicles. J Cell Biol 2021; 219:151777. [PMID: 32406500 PMCID: PMC7300426 DOI: 10.1083/jcb.201806038] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 03/11/2020] [Accepted: 04/24/2020] [Indexed: 02/05/2023] Open
Abstract
Accurate maintenance of organelle identity in the secretory pathway relies on retention and retrieval of resident proteins. In the endoplasmic reticulum (ER), secretory proteins are packaged into COPII vesicles that largely exclude ER residents and misfolded proteins by mechanisms that remain unresolved. Here we combined biochemistry and genetics with correlative light and electron microscopy (CLEM) to explore how selectivity is achieved. Our data suggest that vesicle occupancy contributes to ER retention: in the absence of abundant cargo, nonspecific bulk flow increases. We demonstrate that ER leakage is influenced by vesicle size and cargo occupancy: overexpressing an inert cargo protein or reducing vesicle size restores sorting stringency. We propose that cargo recruitment into vesicles creates a crowded lumen that drives selectivity. Retention of ER residents thus derives in part from the biophysical process of cargo enrichment into a constrained spherical membrane-bound carrier.
Collapse
Affiliation(s)
| | - Alejandro Melero
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Xiao-Han Li
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Jérôme Boulanger
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Wanda Kukulski
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | |
Collapse
|
20
|
Niwa M. A cell cycle checkpoint for the endoplasmic reticulum. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118825. [PMID: 32828757 DOI: 10.1016/j.bbamcr.2020.118825] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
The generation of new cells is one of the most fundamental aspects of cell biology. Proper regulation of the cell cycle is critical for human health, as underscored by many diseases associated with errors in cell cycle regulation, including both cancer and hereditary diseases. A large body of work has identified regulatory mechanisms and checkpoints that ensure accurate and timely replication and segregation of chromosomal DNA. However, few studies have evaluated the extent to which similar checkpoints exist for the division of cytoplasmic components, including organelles. Such checkpoint mechanisms might be crucial for compartments that cannot be generated de novo, such as the endoplasmic reticulum (ER). In this review, we highlight recent work in the model organism Saccharomyces cerevisiae that led to the discovery of such a checkpoint that ensures that cells inherit functional ER into the daughter cell.
Collapse
Affiliation(s)
- Maho Niwa
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, NSB#1, Rm 5328, 9500 Gilman Drive, La Jolla, CA 92093-0377, United States of America.
| |
Collapse
|
21
|
Galeone A, Adams JM, Matsuda S, Presa MF, Pandey A, Han SY, Tachida Y, Hirayama H, Vaccari T, Suzuki T, Lutz CM, Affolter M, Zuberi A, Jafar-Nejad H. Regulation of BMP4/Dpp retrotranslocation and signaling by deglycosylation. eLife 2020; 9:e55596. [PMID: 32720893 PMCID: PMC7394544 DOI: 10.7554/elife.55596] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/23/2020] [Indexed: 12/22/2022] Open
Abstract
During endoplasmic reticulum-associated degradation (ERAD), the cytoplasmic enzyme N-glycanase 1 (NGLY1) is proposed to remove N-glycans from misfolded N-glycoproteins after their retrotranslocation from the ER to the cytosol. We previously reported that NGLY1 regulates Drosophila BMP signaling in a tissue-specific manner (Galeone et al., 2017). Here, we establish the Drosophila Dpp and its mouse ortholog BMP4 as biologically relevant targets of NGLY1 and find, unexpectedly, that NGLY1-mediated deglycosylation of misfolded BMP4 is required for its retrotranslocation. Accumulation of misfolded BMP4 in the ER results in ER stress and prompts the ER recruitment of NGLY1. The ER-associated NGLY1 then deglycosylates misfolded BMP4 molecules to promote their retrotranslocation and proteasomal degradation, thereby allowing properly-folded BMP4 molecules to proceed through the secretory pathway and activate signaling in other cells. Our study redefines the role of NGLY1 during ERAD and suggests that impaired BMP4 signaling might underlie some of the NGLY1 deficiency patient phenotypes.
Collapse
Affiliation(s)
- Antonio Galeone
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Department of Biosciences, University of MilanMilanItaly
| | - Joshua M Adams
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
- Medical Scientist Training Program, Baylor College of MedicineHoustonUnited States
| | | | | | - Ashutosh Pandey
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Seung Yeop Han
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Yuriko Tachida
- Glycometabolome Biochemistry Laboratory, RIKEN Cluster for Pioneering ResearchSaitamaJapan
- T-CiRA joint programKanagawaJapan
| | - Hiroto Hirayama
- Glycometabolome Biochemistry Laboratory, RIKEN Cluster for Pioneering ResearchSaitamaJapan
- T-CiRA joint programKanagawaJapan
| | - Thomas Vaccari
- Department of Biosciences, University of MilanMilanItaly
| | - Tadashi Suzuki
- Glycometabolome Biochemistry Laboratory, RIKEN Cluster for Pioneering ResearchSaitamaJapan
- T-CiRA joint programKanagawaJapan
| | | | | | | | - Hamed Jafar-Nejad
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
22
|
Lajoie P, Snapp EL. Size-dependent secretory protein reflux into the cytosol in association with acute endoplasmic reticulum stress. Traffic 2020; 21:419-429. [PMID: 32246734 PMCID: PMC7317852 DOI: 10.1111/tra.12729] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 01/06/2023]
Abstract
Once secretory proteins have been targeted to the endoplasmic reticulum (ER) lumen, the proteins typically remain partitioned from the cytosol. If the secretory proteins misfold, they can be unfolded and retrotranslocated into the cytosol for destruction by the proteasome by ER-Associated protein Degradation (ERAD). Here, we report that correctly folded and targeted luminal ER fluorescent protein reporters accumulate in the cytosol during acute misfolded secretory protein stress in yeast. Photoactivation fluorescence microscopy experiments reveal that luminal reporters already localized to the ER relocalize to the cytosol, even in the absence of essential ERAD machinery. We named this process "ER reflux." Reflux appears to be regulated in a size-dependent manner for reporters. Interestingly, prior heat shock stress also prevents ER stress-induced reflux. Together, our findings establish a new ER stress-regulated pathway for relocalization of small luminal secretory proteins into the cytosol, distinct from the ERAD and preemptive quality control pathways. Importantly, our results highlight the value of fully characterizing the cell biology of reporters and describe a simple modification to maintain luminal ER reporters in the ER during acute ER stress.
Collapse
Affiliation(s)
- Patrick Lajoie
- Department of Anatomy and Cell BiologyThe University of Western OntarioLondonOntarioCanada
| | | |
Collapse
|
23
|
Kaul Z, Mookherjee D, Das S, Chatterjee D, Chakrabarti S, Chakrabarti O. Loss of tumor susceptibility gene 101 (TSG101) perturbs endoplasmic reticulum structure and function. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118741. [PMID: 32422153 DOI: 10.1016/j.bbamcr.2020.118741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 05/02/2020] [Accepted: 05/08/2020] [Indexed: 12/30/2022]
Abstract
Tumor susceptibility gene 101 (TSG101), an ESCRT-I protein, is implicated in multiple cellular processes and its functional depletion can lead to blocked lysosomal degradation, cell cycle arrest, demyelination and neurodegeneration. Here, we show that loss of TSG101 results in endoplasmic reticulum (ER) stress and this causes ER membrane remodelling (EMR). This correlates with an expansion of ER, increased vacuolation, altered relative distribution of the rough and smooth ER and disruption of three-way junctions. Blocked lysosomal degradation due to TSG101 depletion leads to ER stress and Ca2+ leakage from ER stores, causing destabilization of actin cytoskeleton. Inhibiting Ca2+ release from the ER by blocking ryanodine receptors (RYRs) with Dantrolene partially rescues the ER stress phenotypes. Hence, in this study we have identified the involvement of TSG101 in modulating ER stress mediated remodelling by engaging the actin cytoskeleton. This is significant because functional depletion of TSG101 effectuates ER-stress, perturbs the structure, mobility and function of the ER, all aspects closely associated with neurodegenerative diseases. SUMMARY STATEMENT: We show that tumor susceptibility gene (TSG) 101 regulates endoplasmic reticulum (ER) stress and its membrane remodelling. Loss of TSG101 perturbs structure, mobility and function of the ER as a consequence of actin destabilization.
Collapse
Affiliation(s)
- Zenia Kaul
- Biophysics & Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India; Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA..
| | - Debdatto Mookherjee
- Biophysics & Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Subhrangshu Das
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, CN 6, Sector V, Salt Lake, Kolkata 700091, India
| | - Debmita Chatterjee
- Biophysics & Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Saikat Chakrabarti
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, CN 6, Sector V, Salt Lake, Kolkata 700091, India
| | - Oishee Chakrabarti
- Biophysics & Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India; Homi Bhabha National Institute, India.
| |
Collapse
|
24
|
Tang Q, Zhang X, Cao H, Chen G, Huang H, Zhang P, Zhang Q. A phosphorescent iridium probe for sensing polarity in the endoplasmic reticulum and in vivo. Dalton Trans 2019; 48:7728-7734. [DOI: 10.1039/c9dt01307e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A phosphorescent iridium complex for in situ tracking endoplasmic reticulum polarity variations during ER stress and in vivo.
Collapse
Affiliation(s)
- Qian Tang
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- China
| | - Xuepeng Zhang
- Lab of Computational and Drug Design
- School of Chemical Biology & Biotechnology
- Peking University Shenzhen Graduate School
- Shenzhen
- China
| | - Huiqun Cao
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- China
| | - Ge Chen
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- China
| | - Huaiyi Huang
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-Sen University
- Guangzhou 510275
- China
| | - Pingyu Zhang
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- China
| | - Qianling Zhang
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- China
| |
Collapse
|
25
|
He Y, Shin J, Gong W, Das P, Qu J, Yang Z, Liu W, Kang C, Qu J, Kim JS. Dual-functional fluorescent molecular rotor for endoplasmic reticulum microviscosity imaging during reticulophagy. Chem Commun (Camb) 2019; 55:2453-2456. [DOI: 10.1039/c9cc00300b] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A dual functional fluorescent molecular rotor was developed to trigger intracellular ER autophagy and quantify the local viscosity variations by FLIM imaging.
Collapse
Affiliation(s)
- Ying He
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University
- Shenzhen 518060
- China
| | - Jinwoo Shin
- Department of Chemistry, Korea University
- Seoul 02841
- Korea
| | - Wanjun Gong
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University
- Shenzhen 518060
- China
| | - Pintu Das
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University
- Shenzhen 518060
- China
| | - Jinghan Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University
- Shenzhen 518060
- China
| | - Zhigang Yang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University
- Shenzhen 518060
- China
| | - Wufan Liu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University
- Shenzhen 518060
- China
| | - Chulhun Kang
- Graduate School of East-West Medical Science, Kyung Hee University
- Yongin 446-701
- Korea
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University
- Shenzhen 518060
- China
| | - Jong Seung Kim
- Department of Chemistry, Korea University
- Seoul 02841
- Korea
| |
Collapse
|
26
|
Single particle trajectories reveal active endoplasmic reticulum luminal flow. Nat Cell Biol 2018; 20:1118-1125. [PMID: 30224760 PMCID: PMC6435195 DOI: 10.1038/s41556-018-0192-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 08/09/2018] [Indexed: 01/22/2023]
Abstract
The Endoplasmic Reticulum (ER), a network of membranous sheets and pipes, supports functions encompassing biogenesis of secretory proteins and delivery of functional solutes throughout the cell1,2. Molecular mobility through the ER network enables these functionalities, but diffusion alone is not sufficient to explain luminal transport across supramicron distances. Understanding the ER structure-function relationship is critical in light of mutations in ER morphology regulating proteins that give rise to neurodegenerative disorders3,4. Here, super-resolution microscopy and analysis of single particle trajectories of ER luminal proteins revealed that the topological organization of the ER correlates with distinct trafficking modes of its luminal content: with a dominant diffusive component in tubular junctions and a fast flow component in tubules. Particle trajectory orientations resolved over time revealed an alternating current of the ER contents, whilst fast ER super-resolution identified energy-dependent tubule contraction events at specific points as a plausible mechanism for generating active ER luminal flow. The discovery of active flow in the ER has implications for timely ER content distribution throughout the cell, particularly important for cells with extensive ER-containing projections such as neurons.
Collapse
|
27
|
Lippincott-Schwartz J, Snapp EL, Phair RD. The Development and Enhancement of FRAP as a Key Tool for Investigating Protein Dynamics. Biophys J 2018; 115:1146-1155. [PMID: 30219286 DOI: 10.1016/j.bpj.2018.08.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/27/2018] [Accepted: 08/06/2018] [Indexed: 01/18/2023] Open
Abstract
The saga of fluorescence recovery after photobleaching (FRAP) illustrates how disparate technical developments impact science. Starting with the classic 1976 Axelrod et al. work in Biophysical Journal, FRAP (originally fluorescence photobleaching recovery) opened the door to extraction of quantitative information from photobleaching experiments, laying the experimental and theoretical groundwork for quantifying both the mobility and the mobile fraction of a labeled population of proteins. Over the ensuing years, FRAP's reach dramatically expanded, with new developments in GFP technology and turn-key confocal microscopy, which enabled measurement of protein diffusion and binding/dissociation rates in virtually every compartment within the cell. The FRAP technique and data catalyzed an exchange of ideas between biophysicists studying membrane dynamics, cell biologists focused on intracellular dynamics, and systems biologists modeling the dynamics of cell activity. The outcome transformed the field of cellular biology, leading to a fundamental rethinking of long-held theories of cellular dynamism. Here, we review the pivotal FRAP studies that made these developments and conceptual changes possible, which gave rise to current models of complex cell dynamics.
Collapse
Affiliation(s)
| | - Erik Lee Snapp
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia.
| | - Robert D Phair
- Integrative Bioinformatics, Inc., Mountain View, California
| |
Collapse
|
28
|
Fujii T, Fujita N, Suzuki S, Tsuji T, Takaki T, Umezawa K, Watanabe K, Miyamoto T, Horiuchi K, Matsumoto M, Nakamura M. The unfolded protein response mediated by PERK is casually related to the pathogenesis of intervertebral disc degeneration. J Orthop Res 2018; 36:1334-1345. [PMID: 29080374 DOI: 10.1002/jor.23787] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/26/2017] [Indexed: 02/04/2023]
Abstract
Although the number of patients with intervertebral disc (IVD) degeneration is increasing in aging societies, its etiology and pathogenesis remain elusive and there is currently no effective treatment to prevent this undesirable condition. The unfolded protein response (UPR) is a cellular machinery that plays critical roles in handling endoplasmic reticulum (ER) stress, a condition caused by the accumulation of unfolded proteins in the ER lumen. This study aimed to elucidate the potential role of the UPR mediated by pancreatic endoplasmic reticulum kinase (PERK), one of the major ER stress sensors in mammalian cells, in the development of IVD degeneration. IVD degeneration was artificially induced in Wister rats by percutaneously puncturing the coccyx IVDs and human IVDs were collected from patients who underwent spinal surgery. Expression of the UPR target genes was elevated in degenerative IVDs in both humans and rats. The induction of ER stress in annulus fibrosus cells significantly increased the transcripts for tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) in a nuclear factor (NF)-κB pathway-dependent manner. The expression of TNF-α and IL-6 was significantly reduced by treatment with a selective PERK inhibitor, GSK2606414, and by gene silencing against PERK and activating transcription factor 4 (ATF4) transcripts. Our findings indicate that the UPR mediated by the PERK pathway is causally related to the development of IVD degeneration, suggesting that PERK may be a potential molecular target for suppressing the degenerative changes in IVDs. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1334-1345, 2018.
Collapse
Affiliation(s)
- Takeshi Fujii
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Nobuyuki Fujita
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Satoshi Suzuki
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Takashi Tsuji
- Department of Orthopaedic Surgery, Fujita Health University, Aichi, Japan
| | - Takashi Takaki
- Section of Electron Microscopy, Showa University, Tokyo, Japan.,Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuo Umezawa
- Department of Molecular Target Medicine, Aichi Medical University School of Medicine, Aichi, Japan
| | - Kota Watanabe
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Takeshi Miyamoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Keisuke Horiuchi
- Department of Orthopaedic Surgery, National Defence Medical College, Saitama, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
29
|
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: 39] [Impact Index Per Article: 5.6] [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
|
30
|
Sepulveda D, Rojas-Rivera D, Rodríguez DA, Groenendyk J, Köhler A, Lebeaupin C, Ito S, Urra H, Carreras-Sureda A, Hazari Y, Vasseur-Cognet M, Ali MMU, Chevet E, Campos G, Godoy P, Vaisar T, Bailly-Maitre B, Nagata K, Michalak M, Sierralta J, Hetz C. Interactome Screening Identifies the ER Luminal Chaperone Hsp47 as a Regulator of the Unfolded Protein Response Transducer IRE1α. Mol Cell 2018; 69:238-252.e7. [PMID: 29351844 DOI: 10.1016/j.molcel.2017.12.028] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 10/05/2017] [Accepted: 12/27/2017] [Indexed: 01/17/2023]
Abstract
Maintenance of endoplasmic reticulum (ER) proteostasis is controlled by a dynamic signaling network known as the unfolded protein response (UPR). IRE1α is a major UPR transducer, determining cell fate under ER stress. We used an interactome screening to unveil several regulators of the UPR, highlighting the ER chaperone Hsp47 as the major hit. Cellular and biochemical analysis indicated that Hsp47 instigates IRE1α signaling through a physical interaction. Hsp47 directly binds to the ER luminal domain of IRE1α with high affinity, displacing the negative regulator BiP from the complex to facilitate IRE1α oligomerization. The regulation of IRE1α signaling by Hsp47 is evolutionarily conserved as validated using fly and mouse models of ER stress. Hsp47 deficiency sensitized cells and animals to experimental ER stress, revealing the significance of Hsp47 to global proteostasis maintenance. We conclude that Hsp47 adjusts IRE1α signaling by fine-tuning the threshold to engage an adaptive UPR.
Collapse
Affiliation(s)
- Denisse Sepulveda
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | - Diego Rojas-Rivera
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | - Diego A Rodríguez
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile; Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2S7, Canada
| | - Andres Köhler
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Program of Physiology and Biophysics, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | | | - Shinya Ito
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto and Sangyo University, Motoyama, Kamigamo, Kita-ku, Kyoto 603-8555, Japan
| | - Hery Urra
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | - Amado Carreras-Sureda
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | - Younis Hazari
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | - Mireille Vasseur-Cognet
- Institut d'Ecologie et des Sciences de l'Environnement de Paris, Bondy; Sorbonne Universités, and Institut National de la Santé et de la Recherche Médicale, Paris 7 113, France
| | - Maruf M U Ali
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| | - Eric Chevet
- Inserm U1242, Chemistry, Oncogenesis, Stress, & Signaling, University of Rennes 1, F-35000 Rennes, France; Centre de Lutte le Cancer Eugène Marquis, F-35000 Rennes, France
| | - Gisela Campos
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund 44139, Germany
| | - Patricio Godoy
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund 44139, Germany
| | - Tomas Vaisar
- Division of Metabolism, Endocrinology and Nutrition, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | - Kazuhiro Nagata
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto and Sangyo University, Motoyama, Kamigamo, Kita-ku, Kyoto 603-8555, Japan
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2S7, Canada
| | - Jimena Sierralta
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Program of Physiology and Biophysics, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston MA 02115, USA.
| |
Collapse
|
31
|
Gura Sadovsky R, Brielle S, Kaganovich D, England JL. Measurement of Rapid Protein Diffusion in the Cytoplasm by Photo-Converted Intensity Profile Expansion. Cell Rep 2017; 18:2795-2806. [PMID: 28297680 PMCID: PMC5368347 DOI: 10.1016/j.celrep.2017.02.063] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 12/05/2016] [Accepted: 02/17/2017] [Indexed: 11/27/2022] Open
Abstract
The fluorescence microscopy methods presently used to characterize protein motion in cells infer protein motion from indirect observables, rather than measuring protein motion directly. Operationalizing these methods requires expertise that can constitute a barrier to their broad utilization. Here, we have developed PIPE (photo-converted intensity profile expansion) to directly measure the motion of tagged proteins and quantify it using an effective diffusion coefficient. PIPE works by pulsing photo-convertible fluorescent proteins, generating a peaked fluorescence signal at the pulsed region, and analyzing the spatial expansion of the signal. We demonstrate PIPE's success in measuring accurate diffusion coefficients in silico and in vitro and compare effective diffusion coefficients of native cellular proteins and free fluorophores in vivo. We apply PIPE to measure diffusion anomality in the cell and use it to distinguish free fluorophores from native cellular proteins. PIPE's direct measurement and ease of use make it appealing for cell biologists.
Collapse
Affiliation(s)
- Rotem Gura Sadovsky
- Physics of Living Systems Group, Massachusetts Institute of Technology, Cambridge, MA 02138, USA; Computational and Systems Biology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA 02138, USA
| | - Shlomi Brielle
- Department of Cell and Developmental Biology, Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 91904, Israel; Alexander Grass Center for Bioengineering, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Daniel Kaganovich
- Department of Cell and Developmental Biology, Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 91904, Israel.
| | - Jeremy L England
- Physics of Living Systems Group, Massachusetts Institute of Technology, Cambridge, MA 02138, USA.
| |
Collapse
|
32
|
Xiao H, Wu C, Li P, Gao W, Zhang W, Zhang W, Tong L, Tang B. Ratiometric photoacoustic imaging of endoplasmic reticulum polarity in injured liver tissues of diabetic mice. Chem Sci 2017; 8:7025-7030. [PMID: 29147529 PMCID: PMC5642195 DOI: 10.1039/c7sc02330h] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/07/2017] [Indexed: 01/08/2023] Open
Abstract
As one of the complications of diabetes, liver injury results in significant hazards. Therefore, accurately diagnosing diabetes-induced liver injury beforehand is crucial for the warning and treatment of hepatic diseases. Diabetes-induced liver injury can cause changes in the microstructure and morphology of liver tissue, leading to changes in the hydrophilic and hydrophobic domains in the endoplasmic reticulum (ER), which is closely associated with changes in cellular ER polarity. So, differences in the ER polarity can indicate the degree of diabetes-induced liver injury. Herein, we develop a new fluorescent and photoacoustic dual-mode probe, ER-P, for detection of the ER polarity of liver tissue in normal and diabetic mice. Upon excitation with a 633 nm laser, ER-P showed increasing fluorescence intensity at 800 nm accompanying a decline in the polarity. Due to its polarity-sensitivity, ER-P was utilized for confocal fluorescence imaging in live cells, and the results demonstrate that ER-P can exclusively accumulate in the ER and indicate an increase in the polarity during ER stress. Importantly, ER-P displayed different absorbance intensities at 700 nm and 800 nm in different polarity environments because of intramolecular charge transfer. The photoacoustic intensity ratios between 700 nm and 800 nm will enable quantification of polarity to be achieved. The ratiometric photoacoustic imaging data demonstrate that the polarity of the liver tissue of diabetic mice is higher than that of the liver tissue of normal mice. Meanwhile, after treatment with the antidiabetic drug metformin, diabetic mice exhibit a reduced polarity environment in their liver tissue. The proposed study may serve as a new approach for the early diagnosis and therapeutic evaluation of diabetes-induced liver injury.
Collapse
Affiliation(s)
- Haibin Xiao
- College of Chemistry, Chemical Engineering and Materials Science , Institute of Biomedical Sciences , Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong , Key Laboratory of Molecular and Nano Probes , Ministry of Education , Shandong Normal University , Jinan 250014 , PR China . ;
| | - Chuanchen Wu
- College of Chemistry, Chemical Engineering and Materials Science , Institute of Biomedical Sciences , Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong , Key Laboratory of Molecular and Nano Probes , Ministry of Education , Shandong Normal University , Jinan 250014 , PR China . ;
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science , Institute of Biomedical Sciences , Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong , Key Laboratory of Molecular and Nano Probes , Ministry of Education , Shandong Normal University , Jinan 250014 , PR China . ;
| | - Wen Gao
- College of Chemistry, Chemical Engineering and Materials Science , Institute of Biomedical Sciences , Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong , Key Laboratory of Molecular and Nano Probes , Ministry of Education , Shandong Normal University , Jinan 250014 , PR China . ;
| | - Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science , Institute of Biomedical Sciences , Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong , Key Laboratory of Molecular and Nano Probes , Ministry of Education , Shandong Normal University , Jinan 250014 , PR China . ;
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science , Institute of Biomedical Sciences , Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong , Key Laboratory of Molecular and Nano Probes , Ministry of Education , Shandong Normal University , Jinan 250014 , PR China . ;
| | - Lili Tong
- College of Chemistry, Chemical Engineering and Materials Science , Institute of Biomedical Sciences , Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong , Key Laboratory of Molecular and Nano Probes , Ministry of Education , Shandong Normal University , Jinan 250014 , PR China . ;
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science , Institute of Biomedical Sciences , Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong , Key Laboratory of Molecular and Nano Probes , Ministry of Education , Shandong Normal University , Jinan 250014 , PR China . ;
| |
Collapse
|
33
|
Howell SH. When is the unfolded protein response not the unfolded protein response? PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2017; 260:139-143. [PMID: 28554471 DOI: 10.1016/j.plantsci.2017.03.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/08/2017] [Accepted: 03/18/2017] [Indexed: 05/23/2023]
Abstract
As sessile organisms, plants are subjected to variety of stresses for which they have evolved different protection mechanisms. One mechanism involves endoplasmic reticulum (ER) stress in which the process of protein folding is disturbed and misfolded proteins accumulate in the ER. ER stress elicits the unfolded protein response (UPR) whereby the stress conditions in the ER are communicated to the nucleus to regulate stress response genes. Since the UPR is one of a number of different mechanisms by which plants respond to stress, it is often difficult to distinguish the UPR from other stress responses. Many investigators have relied on the molecular signature of the UPR, the upregulation of UPR genes to implicate the UPR in response to various stresses. However, some of these genes are activated by other stresses making it problematic to know whether the UPR is truly activated in response to a given stress or is part of a complex response. Another challenge is to understand how plants actually perceive different stress conditions. Are all stress conditions that elicit the UPR response caused by an accumulation of misfolded proteins in the ER? Is this the case for salt stress, which induces the UPR? How about biotic stresses, such as bacterial or viral infections? Do they lead to the accumulation of misfolded proteins in the ER or are there other means by which they induce the UPR?
Collapse
Affiliation(s)
- Stephen H Howell
- Plant Sciences Institute and Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
34
|
Hirschhorn T, Levi-Hofman M, Danziger O, Smorodinsky NI, Ehrlich M. Differential molecular regulation of processing and membrane expression of Type-I BMP receptors: implications for signaling. Cell Mol Life Sci 2017; 74:2645-2662. [PMID: 28357470 PMCID: PMC11107780 DOI: 10.1007/s00018-017-2488-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 12/15/2022]
Abstract
The Type-I bone morphogenetic protein receptors (BMPRs), BMPR1A and BMPR1B, present the highest sequence homology among BMPRs, suggestive of functional similitude. However, sequence elements within their extracellular domain, such as signal sequence or N-glycosylation motifs, may result in differential regulation of biosynthetic processing and trafficking and in alterations to receptor function. We show that (i) BMPR1A and the ubiquitous isoform of BMPR1B differed in mode of translocation into the endoplasmic reticulum; and (ii) BMPR1A was N-glycosylated while BMPR1B was not, resulting in greater efficiency of processing and plasma membrane expression of BMPR1A. We further demonstrated the importance of BMPR1A expression and glycosylation in ES-2 ovarian cancer cells, where (i) CRISPR/Cas9-mediated knockout of BMPR1A abrogated BMP2-induced Smad1/5/8 phosphorylation and reduced proliferation of ES-2 cells and (ii) inhibition of N-glycosylation by site-directed mutagenesis, or by tunicamycin or 2-deoxy-D-glucose treatments, reduced biosynthetic processing and plasma membrane expression of BMPR1A and BMP2-induced Smad1/5/8 phosphorylation.
Collapse
Affiliation(s)
- Tal Hirschhorn
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michal Levi-Hofman
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Oded Danziger
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nechama I Smorodinsky
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
35
|
Nixon-Abell J, Obara CJ, Weigel AV, Li D, Legant WR, Xu CS, Pasolli HA, Harvey K, Hess HF, Betzig E, Blackstone C, Lippincott-Schwartz J. Increased spatiotemporal resolution reveals highly dynamic dense tubular matrices in the peripheral ER. Science 2016; 354:aaf3928. [PMID: 27789813 PMCID: PMC6528812 DOI: 10.1126/science.aaf3928] [Citation(s) in RCA: 308] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 09/16/2016] [Indexed: 12/12/2022]
Abstract
The endoplasmic reticulum (ER) is an expansive, membrane-enclosed organelle that plays crucial roles in numerous cellular functions. We used emerging superresolution imaging technologies to clarify the morphology and dynamics of the peripheral ER, which contacts and modulates most other intracellular organelles. Peripheral components of the ER have classically been described as comprising both tubules and flat sheets. We show that this system consists almost exclusively of tubules at varying densities, including structures that we term ER matrices. Conventional optical imaging technologies had led to misidentification of these structures as sheets because of the dense clustering of tubular junctions and a previously uncharacterized rapid form of ER motion. The existence of ER matrices explains previous confounding evidence that had indicated the occurrence of ER "sheet" proliferation after overexpression of tubular junction-forming proteins.
Collapse
Affiliation(s)
- Jonathon Nixon-Abell
- Cell Biology Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, MD, USA. Department of Pharmacology, UCL School of Pharmacy, University College London, London, UK
| | - Christopher J Obara
- Cell Biology and Metabolism Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, USA. Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Aubrey V Weigel
- Cell Biology and Metabolism Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, USA. Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Dong Li
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA. National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Wesley R Legant
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - C Shan Xu
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - H Amalia Pasolli
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Kirsten Harvey
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, UK
| | - Harald F Hess
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Eric Betzig
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Craig Blackstone
- Cell Biology Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, MD, USA.
| | - Jennifer Lippincott-Schwartz
- Cell Biology and Metabolism Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, USA. Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA.
| |
Collapse
|
36
|
Wong AYC, Hristova E, Ahlskog N, Tasse LA, Ngsee JK, Chudalayandi P, Bergeron R. Aberrant Subcellular Dynamics of Sigma-1 Receptor Mutants Underlying Neuromuscular Diseases. Mol Pharmacol 2016; 90:238-53. [PMID: 27418673 DOI: 10.1124/mol.116.104018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/11/2016] [Indexed: 12/16/2023] Open
Abstract
The sigma-1 receptor (σ-1R) is an endoplasmic reticulum resident chaperone protein involved in a plethora of cellular functions, and whose disruption has been implicated in a wide range of diseases. Genetic analysis has revealed two σ-1R mutants involved in neuromuscular disorders. A point mutation (E102Q) in the ligand-binding domain results in the juvenile form of amyotrophic lateral sclerosis (ALS16), and a 20 amino-acid deletion (Δ31-50) in the putative cytosolic domain leads to a form of distal hereditary motor neuropathy. We investigated the localization and functional properties of these mutants in cell lines using confocal imaging and electrophysiology. The σ-1R mutants exhibited a significant increase in mobility, aberrant localization, and enhanced block of the inwardly rectifying K(+) channel Kir2.1, compared with the wild-type σ-1R. Thus, these σ-1R mutants have different functional properties that could contribute to their disease phenotypes.
Collapse
Affiliation(s)
- Adrian Y C Wong
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Elitza Hristova
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Nina Ahlskog
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Louis-Alexandre Tasse
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Johnny K Ngsee
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Prakash Chudalayandi
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Richard Bergeron
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| |
Collapse
|
37
|
Fields PA, Burmester EM, Cox KM, Karch KR. Rapid proteomic responses to a near-lethal heat stress in the salt marsh mussel Geukensia demissa. ACTA ACUST UNITED AC 2016; 219:2673-86. [PMID: 27335449 DOI: 10.1242/jeb.141176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/17/2016] [Indexed: 01/27/2023]
Abstract
Acute heat stress perturbs cellular function on a variety of levels, leading to protein dysfunction and aggregation, oxidative stress and loss of metabolic homeostasis. If these challenges are not overcome quickly, the stressed organism can die. To better understand the earliest tissue-level responses to heat stress, we examined the proteomic response of gill from Geukensia demissa, an extremely eurythermal mussel from the temperate intertidal zone of eastern North America. We exposed 15°C-acclimated individuals to an acute near-lethal heat stress (45°C) for 1 h, and collected gill samples from 0 to 24 h of recovery. The changes in protein expression we found reveal a coordinated physiological response to acute heat stress: proteins associated with apoptotic processes were increased in abundance during the stress itself (i.e. at 0 h of recovery), while protein chaperones and foldases increased in abundance soon after (3 h). The greatest number of proteins changed abundance at 6 h; these included oxidative stress proteins and enzymes of energy metabolism. Proteins associated with the cytoskeleton and extracellular matrix also changed in abundance starting at 6 h, providing evidence of cell proliferation, migration and tissue remodeling. By 12 h, the response to acute heat stress was diminishing, with fewer stress and structural proteins changing in abundance. Finally, the proteins with altered abundances identified at 24 h suggest a return to the pre-stress anabolic state.
Collapse
Affiliation(s)
- Peter A Fields
- Biology Department, Franklin & Marshall College, Lancaster, PA 17603, USA
| | | | - Kelly M Cox
- Biology Department, Franklin & Marshall College, Lancaster, PA 17603, USA
| | - Kelly R Karch
- Biology Department, Franklin & Marshall College, Lancaster, PA 17603, USA
| |
Collapse
|
38
|
Abstract
Transport of newly synthesized proteins from the endoplasmic reticulum (ER) to the Golgi complex is highly selective. As a general rule, such transport is limited to soluble and membrane-associated secretory proteins that have reached properly folded and assembled conformations. To secure the efficiency, fidelity, and control of this crucial transport step, cells use a combination of mechanisms. The mechanisms are based on selective retention of proteins in the ER to prevent uptake into transport vesicles, on selective capture of proteins in COPII carrier vesicles, on inclusion of proteins in these vesicles by default as part of fluid and membrane bulk flow, and on selective retrieval of proteins from post-ER compartments by retrograde vesicle transport.
Collapse
Affiliation(s)
- Charles Barlowe
- Biochemistry Department, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755;
| | - Ari Helenius
- Institute of Biochemistry, ETH Zurich, Zurich CH-8093, Switzerland
| |
Collapse
|
39
|
Piña FJ, Fleming T, Pogliano K, Niwa M. Reticulons Regulate the ER Inheritance Block during ER Stress. Dev Cell 2016; 37:279-88. [PMID: 27117666 DOI: 10.1016/j.devcel.2016.03.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 03/07/2016] [Accepted: 03/28/2016] [Indexed: 01/05/2023]
Abstract
Segregation of functional organelles during the cell cycle is crucial to generate healthy daughter cells. In Saccharomyces cerevisiae, ER stress causes an ER inheritance block to ensure cells inherit a functional ER. Here, we report that formation of tubular ER in the mother cell, the first step in ER inheritance, depends on functional symmetry between the cortical ER (cER) and perinuclear ER (pnER). ER stress induces functional asymmetry, blocking tubular ER formation and ER inheritance. Using fluorescence recovery after photobleaching, we show that the ER chaperone Kar2/BiP fused to GFP and an ER membrane reporter, Hmg1-GFP, behave differently in the cER and pnER. The functional asymmetry and tubular ER formation depend on Reticulons/Yop1, which maintain ER structure. LUNAPARK1 deletion in rtn1Δrtn2Δyop1Δ cells restores the pnER/cER functional asymmetry, tubular ER generation, and ER inheritance blocks. Thus, Reticulon/Yop1-dependent changes in ER structure are linked to ER inheritance during the yeast cell cycle.
Collapse
Affiliation(s)
- Francisco Javier Piña
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, NSB#1, Room 5328, La Jolla, CA 92093-0377, USA
| | - Tinya Fleming
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, NSB#1, Room 4113, La Jolla, CA 92093-0377, USA
| | - Kit Pogliano
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, NSB#1, Room 4113, La Jolla, CA 92093-0377, USA
| | - Maho Niwa
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, NSB#1, Room 5328, La Jolla, CA 92093-0377, USA.
| |
Collapse
|
40
|
Ni Z, Gong Y, Dai X, Ding W, Wang B, Gong H, Qin L, Cheng P, Li S, Lian J, He F. AU4S: a novel synthetic peptide to measure the activity of ATG4 in living cells. Autophagy 2016; 11:403-15. [PMID: 25831015 DOI: 10.1080/15548627.2015.1009773] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
ATG4 plays a key role in autophagy induction, but the methods for monitoring ATG4 activity in living cells are limited. Here we designed a novel fluorescent peptide named AU4S for noninvasive detection of ATG4 activity in living cells, which consists of the cell-penetrating peptide (CPP), ATG4-recognized sequence "GTFG," and the fluorophore FITC. Additionally, an ATG4-resistant peptide AG4R was used as a control. CPP can help AU4S or AG4R to penetrate cell membrane efficiently. AU4S but not AG4R can be recognized and cleaved by ATG4, leading to the change of fluorescence intensity. Therefore, the difference between AU4S- and AG4R-measured fluorescence values in the same sample, defined as "F-D value," can reflect ATG4 activity. By detecting the F-D values, we found that ATG4 activity paralleled LC3B-II levels in rapamycin-treated cells, but neither paralleled LC3B-II levels in starved cells nor presented a correlation with LC3B-II accumulation in WBCs from healthy donors or leukemia patients. However, when DTT was added to the system, ATG4 activity not only paralleled LC3B-II levels in starved cells in the presence or absence of autophagy inhibitors, but also presented a positive correlation with LC3B-II accumulation in WBCs from leukemia patients (R(2) = 0.5288). In conclusion, this study provides a convenient, rapid, and quantitative method to monitor ATG4 activity in living cells, which may be beneficial to basic and clinical research on autophagy.
Collapse
Key Words
- 3-MA, 3-methyladenine
- AG4R, ATG4-resistant peptide
- ATG4
- ATG4, autophagy-related 4, cysteine peptidase
- AU4S
- AU4S, autophagy-related 4 substrate
- Ac, acetyl
- CFP, cyan fluorescent protein
- CPP, cell-penetrating peptide
- CQ, chloroquine
- DTT, dithiothreitol
- EBSS, Earle's balanced salt solution
- FITC, fluorescein isothiocyanate
- HIV, human immunodeficiency virus
- LC3
- MAP1LC3/LC3, microtubule-associated protein 1 light chain 3
- NAC, N-acetyl-L-cysteine
- NRK, normal rat kidney cell line
- PAGE, polyacrylamide gel electrophoresis
- PBS, phosphate-buffered saline
- PE, phosphatidylethanolamine
- PLA2, phospholipase A2
- PMSF, phenylmethanesulfonyl fluoride
- PtdIns3K, phosphatidylinositol 3-kinase
- ROS
- ROS, reactive oxygen species
- SDS, sodium dodecyl sulfate
- WBCs, white blood cells
- YFP, yellow fluorescent protein
- autophagy
Collapse
Affiliation(s)
- Zhenhong Ni
- a Department of Biochemistry and Molecular Biology; College of Basic Medical Sciences ; Third Military Medical University ; Chongqing , China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Protective effect of hyperoside on cardiac ischemia reperfusion injury through inhibition of ER stress and activation of Nrf2 signaling. ASIAN PAC J TROP MED 2016; 9:76-80. [DOI: 10.1016/j.apjtm.2015.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/20/2015] [Accepted: 12/03/2015] [Indexed: 11/20/2022] Open
|
42
|
Tang Y, Li J, Li F, Hu CAA, Liao P, Tan K, Tan B, Xiong X, Liu G, Li T, Yin Y. Autophagy protects intestinal epithelial cells against deoxynivalenol toxicity by alleviating oxidative stress via IKK signaling pathway. Free Radic Biol Med 2015; 89:944-51. [PMID: 26456059 DOI: 10.1016/j.freeradbiomed.2015.09.012] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/12/2015] [Accepted: 09/01/2015] [Indexed: 01/14/2023]
Abstract
Autophagy is an intracellular process of homeostatic degradation that promotes cell survival under various stressors. Deoxynivalenol (DON), a fungal toxin, often causes diarrhea and disturbs the homeostasis of the intestinal system. To investigate the function of intestinal autophagy in response to DON and associated mechanisms, we firstly knocked out ATG5 (autophagy-related gene 5) in porcine intestinal epithelial cells (IPEC-J2) using CRISPR-Cas9 technology. When treated with DON, autophagy was induced in IPEC-J2 cells but not in IPEC-J2.Atg5ko cells. The deficiency in autophagy increased DON-induced apoptosis in IPEC-J2.atg5ko cells, in part, through the generation of reactive oxygen species (ROS). The cellular stress response can be restored in IPEC-J2.atg5ko cells by overexpressing proteins involved in protein folding. Interestingly, we found that autophagy deficiency downregulated the expression of endoplasmic reticulum folding proteins BiP and PDI when IPEC-J2.atg5ko cells were treated with DON. In addition, we investigated the molecular mechanism of autophagy involved in the IKK, AMPK, and mTOR signaling pathway and found that Bay-117082 and Compound C, specific inhibitors for IKK and AMPK, respectively, inhibited the induction of autophagy. Taken together, our results suggest that autophagy is pivotal for protection against DON in pig intestinal cells.
Collapse
Affiliation(s)
- Yulong Tang
- Key Laboratory of Agroecology and Processing of Subtropical Region, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, The Chinese Academy of Science, Ministry of Agriculture, 410125 Changsha City, Hunan, People's Republic of China.
| | - Jianjun Li
- Key Laboratory of Agroecology and Processing of Subtropical Region, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, The Chinese Academy of Science, Ministry of Agriculture, 410125 Changsha City, Hunan, People's Republic of China
| | - Fengna Li
- Key Laboratory of Agroecology and Processing of Subtropical Region, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, The Chinese Academy of Science, Ministry of Agriculture, 410125 Changsha City, Hunan, People's Republic of China.
| | - Chien-An A Hu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131
| | - Peng Liao
- Key Laboratory of Agroecology and Processing of Subtropical Region, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, The Chinese Academy of Science, Ministry of Agriculture, 410125 Changsha City, Hunan, People's Republic of China
| | - Kunrong Tan
- Key Laboratory of Agroecology and Processing of Subtropical Region, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, The Chinese Academy of Science, Ministry of Agriculture, 410125 Changsha City, Hunan, People's Republic of China
| | - Bie Tan
- Key Laboratory of Agroecology and Processing of Subtropical Region, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, The Chinese Academy of Science, Ministry of Agriculture, 410125 Changsha City, Hunan, People's Republic of China
| | - Xia Xiong
- Key Laboratory of Agroecology and Processing of Subtropical Region, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, The Chinese Academy of Science, Ministry of Agriculture, 410125 Changsha City, Hunan, People's Republic of China
| | - Gang Liu
- Key Laboratory of Agroecology and Processing of Subtropical Region, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, The Chinese Academy of Science, Ministry of Agriculture, 410125 Changsha City, Hunan, People's Republic of China
| | - Tiejun Li
- Key Laboratory of Agroecology and Processing of Subtropical Region, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, The Chinese Academy of Science, Ministry of Agriculture, 410125 Changsha City, Hunan, People's Republic of China
| | - Yulong Yin
- Key Laboratory of Agroecology and Processing of Subtropical Region, Institute of Subtropical Agriculture, Research Center for Healthy Breeding of Livestock and Poultry, Hunan Engineering and Research Center for Animal and Poultry Science, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central China, The Chinese Academy of Science, Ministry of Agriculture, 410125 Changsha City, Hunan, People's Republic of China; School of Life Sciences, Hunan Normal University, Changsha 41008, China
| |
Collapse
|
43
|
Piña FJ, Niwa M. The ER Stress Surveillance (ERSU) pathway regulates daughter cell ER protein aggregate inheritance. eLife 2015; 4. [PMID: 26327697 PMCID: PMC4555637 DOI: 10.7554/elife.06970] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 08/04/2015] [Indexed: 12/11/2022] Open
Abstract
Stress induced by cytoplasmic protein aggregates can have deleterious consequences for the cell, contributing to neurodegeneration and other diseases. Protein aggregates are also formed within the endoplasmic reticulum (ER), although the fate of ER protein aggregates, specifically during cell division, is not well understood. By simultaneous visualization of both the ER itself and ER protein aggregates, we found that ER protein aggregates that induce ER stress are retained in the mother cell by activation of the ER Stress Surveillance (ERSU) pathway, which prevents inheritance of stressed ER. In contrast, under conditions of normal ER inheritance, ER protein aggregates can enter the daughter cell. Thus, whereas cytoplasmic protein aggregates are retained in the mother cell to protect the functional capacity of daughter cells, the fate of ER protein aggregates is determined by whether or not they activate the ERSU pathway to impede transmission of the cortical ER during the cell cycle.
Collapse
Affiliation(s)
- Francisco J Piña
- Division of Biological Sciences, Section of Molecular Biology, Univeristy of California, San Diego, San Diego, United States
| | - Maho Niwa
- Division of Biological Sciences, Section of Molecular Biology, Univeristy of California, San Diego, San Diego, United States
| |
Collapse
|
44
|
Wei X, Howell AS, Dong X, Taylor CA, Cooper RC, Zhang J, Zou W, Sherwood DR, Shen K. The unfolded protein response is required for dendrite morphogenesis. eLife 2015; 4:e06963. [PMID: 26052671 PMCID: PMC4484204 DOI: 10.7554/elife.06963] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 06/07/2015] [Indexed: 11/13/2022] Open
Abstract
Precise patterning of dendritic fields is essential for the formation and function of neuronal circuits. During development, dendrites acquire their morphology by exuberant branching. How neurons cope with the increased load of protein production required for this rapid growth is poorly understood. Here we show that the physiological unfolded protein response (UPR) is induced in the highly branched Caenorhabditis elegans sensory neuron PVD during dendrite morphogenesis. Perturbation of the IRE1 arm of the UPR pathway causes loss of dendritic branches, a phenotype that can be rescued by overexpression of the ER chaperone HSP-4 (a homolog of mammalian BiP/ grp78). Surprisingly, a single transmembrane leucine-rich repeat protein, DMA-1, plays a major role in the induction of the UPR and the dendritic phenotype in the UPR mutants. These findings reveal a significant role for the physiological UPR in the maintenance of ER homeostasis during morphogenesis of large dendritic arbors. DOI:http://dx.doi.org/10.7554/eLife.06963.001 The brain consists of billions of cells called neurons that can rapidly send and receive information. At one end of the neuron, branched structures called dendrites receive signals from other cells. The number of dendrites and the amount of branching vary in different types of neurons. These patterns are crucial for each neuron to receive the information it needs. Abnormalities in dendrites affect brain activity and are associated with several diseases in humans. To make dendrites, the neuron needs to increase the amount of protein and other cell materials it produces. New proteins are made in a compartment called the endoplasmic reticulum and are folded into particular three-dimensional shapes with the help of chaperone proteins. These chaperones may be overwhelmed if protein production increases, leading to some proteins being folded incorrectly. This can activate a system called the unfolded protein response, which increases the number of chaperone proteins so that the proteins can be refolded correctly. However, it was not clear if neurons rely on the unfolded protein response, or another system, to cope with the increased levels of protein production needed to form complicated dendrite structures. Wei et al. studied a type of neuron called PVD—which has an elaborate network of dendrites—in nematode worms. The experiments show that the unfolded protein response is activated in these neurons as the dendrites form. Mutant worms that were missing a protein called IRE1, which can activate the unfolded protein response, had dendrites with fewer branches than normal worms. The experiments also show that a protein called DMA-1—which is required for dendrites to form—was not able to fold correctly in the mutant worms. As a result, this protein remained in the endoplasmic reticulum instead of moving to the surface of the cell where it is usually found. Wei et al.'s findings reveal that the unfolded protein response plays a major role in allowing cells to increase protein production as the dendrites form. The next challenge is to understand how neurons coordinate transcription and activation of the unfolded protein response. DOI:http://dx.doi.org/10.7554/eLife.06963.002
Collapse
Affiliation(s)
- Xing Wei
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Audrey S Howell
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Xintong Dong
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Caitlin A Taylor
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Roshni C Cooper
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Jianqi Zhang
- Division of Biostatistics, Department of Preventive Medicine, University of Southern California, Los Angeles, United States
| | - Wei Zou
- Department of Biology, Duke University, Durham, United States
| | | | - Kang Shen
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| |
Collapse
|
45
|
Yang Z, He Y, Lee JH, Chae WS, Ren WX, Lee JH, Kang C, Kim JS. A Nile Red/BODIPY-based bimodal probe sensitive to changes in the micropolarity and microviscosity of the endoplasmic reticulum. Chem Commun (Camb) 2015; 50:11672-5. [PMID: 25140835 DOI: 10.1039/c4cc04915b] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We herein report a fluorescent bimodal probe (1) capable of determining ER viscosity and polarity changes using FLIM and fluorescence ratiometry, respectively; during ER stress caused by tunicamycin, the viscosity was increased from ca. 129.5 to 182.0 cP and the polarity of the ER (dielectric constant, ε) enhanced from 18.5 to 21.1.
Collapse
Affiliation(s)
- Zhigang Yang
- Department of Chemistry, Korea University, Seoul 136-701, Korea.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Li GY, Fan B, Jiao YY. Rapamycin attenuates visible light-induced injury in retinal photoreceptor cells via inhibiting endoplasmic reticulum stress. Brain Res 2014; 1563:1-12. [PMID: 24607296 DOI: 10.1016/j.brainres.2014.02.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/19/2014] [Accepted: 02/08/2014] [Indexed: 01/08/2023]
Abstract
An extended exposure of the retina to visible light may lead to photochemical damage in retinal photoreceptor cells. The exact mechanism of retinal light damage remains unknown, and an effective therapy is still unavailable. Here, we demonstrated that rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR), markedly protected 661W photoreceptor cells from visible light exposure-induced damage at the nanomolar level. We also observed by transmission electron microscopy that light exposure led to severe endoplasmic reticulum (ER) stress in 661W cells as well as abnormal endomembranes and ER membranes. In addition, obvious upregulated ER stress markers were monitored by western blot at the protein level and by quantitative reverse transcription-polymerase chain reaction (RT-PCR) at the mRNA level. Interestingly, rapamycin pretreatment significantly suppressed light-induced ER stress and all three major branches of the unfolded protein response (UPR), including the RNA-dependent protein kinase-like ER kinase (PERK), inositol-requiring enzyme 1 (IRE1), and activating transcription factor 6 (ATF6) pathways both at the protein and mRNA levels. Additionally, the inhibition of ER stress by rapamycin was further confirmed with a dithiothreitol (DTT; a classical ER stress inducer)-damaged 661W cell model. Meanwhile, our results also revealed that rapamycin was able to remarkably inhibit the activation of mTOR and its downstream factors eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1), p-4EBP1, p70, p-p70, and phosphorylated ribosomal protein S6 kinase (p-S6K) in the light-injured 661W cells. Thus, these data indicate that visible light induces ER stress in 661W cells; whereas the mTOR inhibitor, rapamycin, effectively protects 661W cells from light injury through suppressing the ER stress pathway.
Collapse
Affiliation(s)
- Guang-Yu Li
- Department of Ophthalmology, Second Hospital of JiLin University, ChangChun 130041, China.
| | - Bin Fan
- Department of Ophthalmology, Second Hospital of JiLin University, ChangChun 130041, China.
| | - Ying-Ying Jiao
- Department of Ophthalmology, Second Hospital of JiLin University, ChangChun 130041, China
| |
Collapse
|
47
|
Approaches to imaging unfolded secretory protein stress in living cells. ENDOPLASMIC RETICULUM STRESS IN DISEASES 2014; 1:27-39. [PMID: 25419521 DOI: 10.2478/ersc-2014-0002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The endoplasmic reticulum (ER) is the point of entry of proteins into the secretory pathway. Nascent peptides interact with the ER quality control machinery that ensures correct folding of the nascent proteins. Failure to properly fold proteins can lead to loss of protein function and cytotoxic aggregation of misfolded proteins that can lead to cell death. To cope with increases in the ER unfolded secretory protein burden, cells have evolved the Unfolded Protein Response (UPR). The UPR is the primary signaling pathway that monitors the state of the ER folding environment. When the unfolded protein burden overwhelms the capacity of the ER quality control machinery, a state termed ER stress, sensor proteins detect accumulation of misfolded peptides and trigger the UPR transcriptional response. The UPR, which is conserved from yeast to mammals, consists of an ensemble of complex signaling pathways that aims at adapting the ER to the new misfolded protein load. To determine how different factors impact the ER folding environment, various tools and assays have been developed. In this review, we discuss recent advances in live cell imaging reporters and model systems that enable researchers to monitor changes in the unfolded secretory protein burden and activation of the UPR and its associated signaling pathways.
Collapse
|
48
|
Ambrose RL, Mackenzie JM. Flaviviral regulation of the unfolded protein response: can stress be beneficial? Future Virol 2013. [DOI: 10.2217/fvl.13.100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Members of the Flaviviridae family remain some of the most significant human viral pathogens, with few vaccines or antivirals commercially available for therapeutic use. Thus, understanding the intracellular events of replication and how these viruses modulate signaling within an infected cell is of great importance. The ER is central to replication within the Flaviviridae family, as the site of viral protein translation and processing, as a source of membranes for replication complex formation and as a site of virus assembly. This places a large burden upon the organelle, resulting in the induction of ER stress responses, in particular the unfolded protein response. In turn, unfolded protein response signaling induced in infected cells is tightly modulated by the virus in order to maintain an optimal environment for replication. The loss of various components of the stress response can have either beneficial or detrimental effects, presenting intriguing targets for antiviral discovery.
Collapse
Affiliation(s)
- Rebecca L Ambrose
- Department of Microbiology & Immunology, University of Melbourne, Parkville, Melbourne, VIC, 3052, Australia
| | - Jason M Mackenzie
- Department of Microbiology & Immunology, University of Melbourne, Parkville, Melbourne, VIC, 3052, Australia
| |
Collapse
|
49
|
Gidalevitz T, Stevens F, Argon Y. Orchestration of secretory protein folding by ER chaperones. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1833:2410-24. [PMID: 23507200 PMCID: PMC3729627 DOI: 10.1016/j.bbamcr.2013.03.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 02/27/2013] [Accepted: 03/01/2013] [Indexed: 02/06/2023]
Abstract
The endoplasmic reticulum is a major compartment of protein biogenesis in the cell, dedicated to production of secretory, membrane and organelle proteins. The secretome has distinct structural and post-translational characteristics, since folding in the ER occurs in an environment that is distinct in terms of its ionic composition, dynamics and requirements for quality control. The folding machinery in the ER therefore includes chaperones and folding enzymes that introduce, monitor and react to disulfide bonds, glycans, and fluctuations of luminal calcium. We describe the major chaperone networks in the lumen and discuss how they have distinct modes of operation that enable cells to accomplish highly efficient production of the secretome. This article is part of a Special Issue entitled: Functional and structural diversity of endoplasmic reticulum.
Collapse
Affiliation(s)
- Tali Gidalevitz
- Department of Biology, Drexel University, Drexel University, 418 Papadakis Integrated Science Bldg, 3245 Chestnut Street, Philadelphia, PA 19104
| | | | - Yair Argon
- Division of Cell Pathology, Department of Pathology and Lab Medicine, The Children's Hospital of Philadelphia and the University of Pennsylvania, 3615 Civic Center Blvd., Philadelphia, PA 19104, USA, , Phone: 267-426-5131, Fax: 267-426-5165)
| |
Collapse
|
50
|
Costantini L, Snapp E. Probing endoplasmic reticulum dynamics using fluorescence imaging and photobleaching techniques. CURRENT PROTOCOLS IN CELL BIOLOGY 2013; 60:21.7.1-21.7.29. [PMID: 24510787 PMCID: PMC3920296 DOI: 10.1002/0471143030.cb2107s60] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This unit describes approaches and tools for studying the dynamics and organization of endoplasmic reticulum (ER) membranes and proteins in living cells using fluorescence microscopy. The ER plays a key role in secretory protein biogenesis, calcium regulation, and lipid synthesis. However, study of these processes has often been restricted to biochemical assays that average millions of lysed cells or imaging of static fixed cells. With new fluorescent protein (FP) reporter tools, sensitive commercial microscopes, and photobleaching techniques, investigators can interrogate the behaviors of ER proteins, membranes, and stress pathways in single live cells. Solutions are described for imaging challenges relevant to the ER, including the mobility of ER membranes, a range of ER structures, and the influence of post-translational modifications on FP reporters. Considerations for performing photobleaching assays for ER proteins are discussed. Finally, reporters and drugs for studying misfolded secretory protein stress and the unfolded protein response are described.
Collapse
Affiliation(s)
- Lindsey Costantini
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| | - Erik Snapp
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| |
Collapse
|