1
|
Xu S, Xiao S, Qi J, Yao M, He P, Wang R, Wei E, Wang Q, Zhang Y, Tang X, Shen Z. Glucose-regulated protein 78 regulates the subunit-folding of the CCT complex by modulating gene expression and protein interaction in the microsporidian Nosema bombycis. Int J Biol Macromol 2025; 290:138971. [PMID: 39708871 DOI: 10.1016/j.ijbiomac.2024.138971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Chaperonin containing tailless complex polypeptide 1 (CCT) functions as a molecular chaperone and is essential for ensuring proper protein folding. Glucose-regulated protein 78 (GRP78/Bip), also a type of chaperone, not only assists in folding of proteins, but also facilitates the transportation of proteins into the endoplasmic reticulum (ER) via the Sec protein complex. In this study, we identified the CCTη of N. bombycis (NbCCTη) for the first time. Immunoprecipitations and mass spectrometry (IP-MS) of NbCCTη analysis showed that NbBip may interact with CCT subunits. Yeast two-hybrid assays validated that NbBip interacts with NbCCTη, as well as NbCCTα and NbCCTε. Furthermore, RNA interference on NbBip brought about radical expression of NbCCTα, NbCCTε, and NbCCTη, while RNAi on NbCCT subunits resulted in abnormal expression of NbBip. Immunofluorescence assay results showed that NbBip colocalized with NbCCTα and NbCCTη, and CCTη colocalized with Nbβ-tubulin and Nbactin in the parasite. Collectively, these findings suggest that NbBip may act as a crucial factor in the subunit-folding and assembly of CCT complex in N. bombycis.
Collapse
Affiliation(s)
- Sheng Xu
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Shengyan Xiao
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Jingru Qi
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Mingshuai Yao
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Ping He
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Runpeng Wang
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Erjun Wei
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Qiang Wang
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China; Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, People's Republic of China
| | - Yiling Zhang
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China; Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, People's Republic of China
| | - Xudong Tang
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China; Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, People's Republic of China
| | - Zhongyuan Shen
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China; Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, People's Republic of China.
| |
Collapse
|
2
|
Bhakta K, Roy M, Samanta S, Ghosh A. Functional diversity in archaeal Hsp60: a molecular mosaic of Group I and Group II chaperonin. FEBS J 2024; 291:4323-4348. [PMID: 38923213 DOI: 10.1111/febs.17213] [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: 02/19/2024] [Revised: 05/23/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
External stress disrupts the balance of protein homeostasis, necessitating the involvement of heat shock proteins (Hsps) in restoring equilibrium and ensuring cellular survival. The thermoacidophilic crenarchaeon Sulfolobus acidocaldarius, lacks the conventional Hsp100, Hsp90, and Hsp70, relying solely on a single ATP-dependent Group II chaperonin, Hsp60, comprising three distinct subunits (α, β, and γ) to refold unfolded substrates and maintain protein homeostasis. Hsp60 forms three different complexes, namely Hsp60αβγ, Hsp60αβ, and Hsp60β, at temperatures of 60 °C, 75 °C, and 90 °C, respectively. This study delves into the intricacies of Hsp60 complexes in S. acidocaldarius, uncovering their ability to form oligomeric structures in the presence of ATP. The recognition of substrates by Hsp60 involves hydrophobic interactions, and the subsequent refolding process occurs in an ATP-dependent manner through charge-driven interactions. Furthermore, the Hsp60β homo-oligomeric complex can protect the archaeal and eukaryotic membrane from stress-induced damage. Hsp60 demonstrates nested cooperativity in ATP hydrolysis activity, where MWC-type cooperativity is nested within KNF-type cooperativity. Remarkably, during ATP hydrolysis, Hsp60β, and Hsp60αβ complexes exhibit a mosaic behavior, aligning with characteristics observed in both Group I and Group II chaperonins, adding a layer of complexity to their functionality.
Collapse
Affiliation(s)
- Koustav Bhakta
- Department of Biological Sciences, Bose Institute, Kolkata, India
| | - Mousam Roy
- Department of Biological Sciences, Bose Institute, Kolkata, India
| | - Shirsha Samanta
- Department of Biological Sciences, Bose Institute, Kolkata, India
| | - Abhrajyoti Ghosh
- Department of Biological Sciences, Bose Institute, Kolkata, India
| |
Collapse
|
3
|
Gu Y, Pope A, Smith C, Carmona C, Johnstone A, Shi L, Chen X, Santos S, Bacon-Brenes CC, Shoff T, Kleczko KM, Frydman J, Thompson LM, Mobley WC, Wu C. BDNF and TRiC-inspired reagent rescue cortical synaptic deficits in a mouse model of Huntington's disease. Neurobiol Dis 2024; 195:106502. [PMID: 38608784 PMCID: PMC11890210 DOI: 10.1016/j.nbd.2024.106502] [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: 01/04/2024] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024] Open
Abstract
Synaptic changes are early manifestations of neuronal dysfunction in Huntington's disease (HD). However, the mechanisms by which mutant HTT protein impacts synaptogenesis and function are not well understood. Herein we explored HD pathogenesis in the BACHD mouse model by examining synaptogenesis and function in long term primary cortical cultures. At DIV14 (days in vitro), BACHD cortical neurons showed no difference from WT neurons in synaptogenesis as revealed by colocalization of a pre-synaptic (Synapsin I) and a post-synaptic (PSD95) marker. From DIV21 to DIV35, BACHD neurons showed progressively reduced colocalization of Synapsin I and PSD95 relative to WT neurons. The deficits were effectively rescued by treatment of BACHD neurons with BDNF. The recombinant apical domain of CCT1 (ApiCCT1) yielded a partial rescuing effect. BACHD neurons also showed culture age-related significant functional deficits as revealed by multielectrode arrays (MEAs). These deficits were prevented by BDNF, whereas ApiCCT1 showed a less potent effect. These findings are evidence that deficits in BACHD synapse and function can be replicated in vitro and that BDNF or a TRiC-inspired reagent can potentially be protective against these changes in BACHD neurons. Our findings support the use of cellular models to further explicate HD pathogenesis and potential treatments.
Collapse
Affiliation(s)
- Yingli Gu
- Department of Neurology, The Fourth Hospital of Harbin Medical University, 150001, China; Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Alexander Pope
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Charlene Smith
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, United States of America
| | - Christopher Carmona
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America; Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, United States of America; Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, United States of America; Beckman Laser Institute & Medical Clinic, University of California, Irvine, Irvine, CA, United States; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Aaron Johnstone
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Linda Shi
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, United States of America; Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, United States of America; Beckman Laser Institute & Medical Clinic, University of California, Irvine, Irvine, CA, United States; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Xuqiao Chen
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Sarai Santos
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | | | - Thomas Shoff
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Korbin M Kleczko
- Department of Biology and Genetics, Stanford University, Stanford, CA 94305-5430, United States of America
| | - Judith Frydman
- Department of Biology and Genetics, Stanford University, Stanford, CA 94305-5430, United States of America
| | - Leslie M Thompson
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, United States of America; Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, United States of America; Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States of America; Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697, United States of America
| | - William C Mobley
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America.
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America.
| |
Collapse
|
4
|
Pinho-Correia LM, Prokop A. Maintaining essential microtubule bundles in meter-long axons: a role for local tubulin biogenesis? Brain Res Bull 2023; 193:131-145. [PMID: 36535305 DOI: 10.1016/j.brainresbull.2022.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Axons are the narrow, up-to-meter long cellular processes of neurons that form the biological cables wiring our nervous system. Most axons must survive for an organism's lifetime, i.e. up to a century in humans. Axonal maintenance depends on loose bundles of microtubules that run without interruption all along axons. The continued turn-over and the extension of microtubule bundles during developmental, regenerative or plastic growth requires the availability of α/β-tubulin heterodimers up to a meter away from the cell body. The underlying regulation in axons is poorly understood and hardly features in past and contemporary research. Here we discuss potential mechanisms, particularly focussing on the possibility of local tubulin biogenesis in axons. Current knowledge might suggest that local translation of tubulin takes place in axons, but far less is known about the post-translational machinery of tubulin biogenesis involving three chaperone complexes: prefoldin, CCT and TBC. We discuss functional understanding of these chaperones from a range of model organisms including yeast, plants, flies and mice, and explain what is known from human diseases. Microtubules across species depend on these chaperones, and they are clearly required in the nervous system. However, most chaperones display a high degree of functional pleiotropy, partly through independent functions of individual subunits outside their complexes, thus posing a challenge to experimental studies. Notably, we found hardly any studies that investigate their presence and function particularly in axons, thus highlighting an important gap in our understanding of axon biology and pathology.
Collapse
Affiliation(s)
- Liliana Maria Pinho-Correia
- The University of Manchester, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biology, Manchester, UK
| | - Andreas Prokop
- The University of Manchester, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biology, Manchester, UK.
| |
Collapse
|
5
|
Li M, Zeng J, Chang Y, Lv L, Ye G. CCT3 as a Diagnostic and Prognostic Biomarker in Cervical Cancer. Crit Rev Eukaryot Gene Expr 2023; 33:17-28. [PMID: 37522542 DOI: 10.1615/critreveukaryotgeneexpr.2023048208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
The chaperonin-containing TCP1 complex subunit 3 (CCT3) has been reported to be involved in the development and prognosis of many tumors, including cervical cancer (CC). This study aimed to analyze the expression and prognostic value of CCT3 in CC by bioinformatics and retrospective study. CCT3 gene expression profiles and clinical information in CC were downloaded from the cancer genome atlas (TCGA) and gene expression omnibus (GEO) databases. CCT3 expression was verified by quantitative real-time polymerase chain reaction (RT-qPCR), Western blot, and immunohistochemistry (IHC). Logistic regression and chi-square testing were used to analyze the relationship between CCT3 expression and the clinical characteristics of CC. Kaplan-Meier and Cox analyses were used to evaluate whether CCT3 affects the prognosis of CC. Nomogram and calibration curves were used to test the predictive value of CCT3. The expression of CCT3 in CC tissues was significantly upregulated compared with that in adjacent benign tissues, and was related to HPV16/18 infection, grade, and positive lymph nodes. High expression of CCT3 is associated with poor prognosis of CC and can be used as an independent risk factor for CC. The prognostic model based on CCT3 and CC clinical features has good predictive ability. CCT3 is overexpressed in CC, which is related to poor prognosis and expected to become a biomarker for CC.
Collapse
Affiliation(s)
- Man Li
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233030, China
| | - Jianmin Zeng
- Affiliated Hospital of Kunming University of Science and Technology, First People's Hospital of Yunnan Province, Kunming, 650500, China
| | - Yuhuan Chang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233030, China
| | - Lili Lv
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233030, China
| | - Guoliu Ye
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233030, China
| |
Collapse
|
6
|
Smith BJ, Brandão-Teles C, Zuccoli GS, Reis-de-Oliveira G, Fioramonte M, Saia-Cereda VM, Martins-de-Souza D. Protein Succinylation and Malonylation as Potential Biomarkers in Schizophrenia. J Pers Med 2022; 12:jpm12091408. [PMID: 36143193 PMCID: PMC9500613 DOI: 10.3390/jpm12091408] [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: 07/05/2022] [Revised: 08/24/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Two protein post-translational modifications, lysine succinylation and malonylation, are implicated in protein regulation, glycolysis, and energy metabolism. The precursors of these modifications, succinyl-CoA and malonyl-CoA, are key players in central metabolic processes. Both modification profiles have been proven to be responsive to metabolic stimuli, such as hypoxia. As mitochondrial dysfunction and metabolic dysregulation are implicated in schizophrenia and other psychiatric illnesses, these modification profiles have the potential to reveal yet another layer of protein regulation and can furthermore represent targets for biomarkers that are indicative of disease as well as its progression and treatment. In this work, data from shotgun mass spectrometry-based quantitative proteomics were compiled and analyzed to probe the succinylome and malonylome of postmortem brain tissue from patients with schizophrenia against controls and the human oligodendrocyte precursor cell line MO3.13 with the dizocilpine chemical model for schizophrenia, three antipsychotics, and co-treatments. Several changes in the succinylome and malonylome were seen in these comparisons, revealing these modifications to be a largely under-studied yet important form of protein regulation with broad potential applications.
Collapse
Affiliation(s)
- Bradley Joseph Smith
- Laboratory of Neuroproteomics, Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas 13083-862, Brazil
- Correspondence: (B.J.S.); (D.M.-d.-S.); Tel.: +55-(19)-3521-6129 (D.M.-d.-S.)
| | - Caroline Brandão-Teles
- Laboratory of Neuroproteomics, Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas 13083-862, Brazil
| | - Giuliana S. Zuccoli
- Laboratory of Neuroproteomics, Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas 13083-862, Brazil
| | - Guilherme Reis-de-Oliveira
- Laboratory of Neuroproteomics, Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas 13083-862, Brazil
| | - Mariana Fioramonte
- Laboratory of Neuroproteomics, Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas 13083-862, Brazil
| | - Verônica M. Saia-Cereda
- Laboratory of Neuroproteomics, Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas 13083-862, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas 13083-862, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo 05403-000, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas 13083-862, Brazil
- D’Or Institute for Research and Education (IDOR), São Paulo 04501-000, Brazil
- Correspondence: (B.J.S.); (D.M.-d.-S.); Tel.: +55-(19)-3521-6129 (D.M.-d.-S.)
| |
Collapse
|
7
|
Roy M, Bhakta K, Ghosh A. Minimal Yet Powerful: The Role of Archaeal Small Heat Shock Proteins in Maintaining Protein Homeostasis. Front Mol Biosci 2022; 9:832160. [PMID: 35647036 PMCID: PMC9133787 DOI: 10.3389/fmolb.2022.832160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/02/2022] [Indexed: 11/21/2022] Open
Abstract
Small heat shock proteins (sHsp) are a ubiquitous group of ATP-independent chaperones found in all three domains of life. Although sHsps in bacteria and eukaryotes have been studied extensively, little information was available on their archaeal homologs until recently. Interestingly, archaeal heat shock machinery is strikingly simplified, offering a minimal repertoire of heat shock proteins to mitigate heat stress. sHsps play a crucial role in preventing protein aggregation and holding unfolded protein substrates in a folding-competent form. Besides protein aggregation protection, archaeal sHsps have been shown recently to stabilize membranes and contribute to transferring captured substrate proteins to chaperonin for refolding. Furthermore, recent studies on archaeal sHsps have shown that environment-induced oligomeric plasticity plays a crucial role in maintaining their functional form. Despite being prokaryotes, the archaeal heat shock protein repository shares several features with its highly sophisticated eukaryotic counterpart. The minimal nature of the archaeal heat shock protein repository offers ample scope to explore the function and regulation of heat shock protein(s) to shed light on their evolution. Moreover, similar structural dynamics of archaeal and human sHsps have made the former an excellent system to study different chaperonopathies since archaeal sHsps are more stable under in vitro experiments.
Collapse
|
8
|
Tahmaz I, Shahmoradi Ghahe S, Topf U. Prefoldin Function in Cellular Protein Homeostasis and Human Diseases. Front Cell Dev Biol 2022; 9:816214. [PMID: 35111762 PMCID: PMC8801880 DOI: 10.3389/fcell.2021.816214] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/29/2021] [Indexed: 01/05/2023] Open
Abstract
Cellular functions are largely performed by proteins. Defects in the production, folding, or removal of proteins from the cell lead to perturbations in cellular functions that can result in pathological conditions for the organism. In cells, molecular chaperones are part of a network of surveillance mechanisms that maintains a functional proteome. Chaperones are involved in the folding of newly synthesized polypeptides and assist in refolding misfolded proteins and guiding proteins for degradation. The present review focuses on the molecular co-chaperone prefoldin. Its canonical function in eukaryotes involves the transfer of newly synthesized polypeptides of cytoskeletal proteins to the tailless complex polypeptide 1 ring complex (TRiC/CCT) chaperonin which assists folding of the polypeptide chain in an energy-dependent manner. The canonical function of prefoldin is well established, but recent research suggests its broader function in the maintenance of protein homeostasis under physiological and pathological conditions. Interestingly, non-canonical functions were identified for the prefoldin complex and also for its individual subunits. We discuss the latest findings on the prefoldin complex and its subunits in the regulation of transcription and proteasome-dependent protein degradation and its role in neurological diseases, cancer, viral infections and rare anomalies.
Collapse
Affiliation(s)
- Ismail Tahmaz
- Laboratory of Molecular Basis of Aging and Rejuvenation, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Somayeh Shahmoradi Ghahe
- Laboratory of Molecular Basis of Aging and Rejuvenation, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Ulrike Topf
- Laboratory of Molecular Basis of Aging and Rejuvenation, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
9
|
Roy M, Bhakta K, Bhowmick A, Gupta S, Ghosh A, Ghosh A. Archaeal Hsp14 drives substrate shuttling between small heat shock proteins and thermosome: insights into a novel substrate transfer pathway. FEBS J 2021; 289:1080-1104. [PMID: 34637594 DOI: 10.1111/febs.16226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/26/2021] [Accepted: 10/11/2021] [Indexed: 12/25/2022]
Abstract
Heat shock proteins maintain protein homeostasis and facilitate the survival of an organism under stress. Archaeal heat shock machinery usually consists of only sHsps, Hsp70, and Hsp60. Moreover, Hsp70 is absent in thermophilic and hyperthermophilic archaea. In the absence of Hsp70, how aggregating protein substrates are transferred to Hsp60 for refolding remains elusive. Here, we investigated the crosstalk in the heat shock response pathway of thermoacidophilic crenarchaeon Sulfolobus acidocaldarius. In the present study, we biophysically and biochemically characterized one of the small heat shock proteins, Hsp14, of S. acidocaldarius. Moreover, we investigated its ability to interact with Hsp20 and Hsp60 to facilitate the substrate proteins' folding under stress conditions. Like Hsp20, we demonstrated that the dimer is the active form of Hsp14, and it forms an oligomeric storage form at a higher temperature. More importantly, the dynamics of the Hsp14 oligomer are maintained by rapid subunit exchange between the dimeric states, and the rate of subunit exchange increases with increasing temperature. We also tested the ability of Hsp14 to form hetero-oligomers via subunit exchange with Hsp20. We observed hetero-oligomer formation only at higher temperatures (50 °C-70 °C). Furthermore, experiments were performed to investigate the interaction between small heat shock proteins and Hsp60. We demonstrated an enthalpy-driven direct physical interaction between Hsp14 and Hsp60. Our results revealed that Hsp14 could transfer sHsp-captured substrate proteins to Hsp60, which then refolds them back to their active form.
Collapse
Affiliation(s)
- Mousam Roy
- Department of Biochemistry, Bose Institute, Kolkata, India
| | - Koustav Bhakta
- Department of Biochemistry, Bose Institute, Kolkata, India
| | | | | | - Anupama Ghosh
- Division of Plant Biology, Bose Institute, Kolkata, India
| | | |
Collapse
|
10
|
Chaperonin point mutation enhances cadmium endurance in Saccharomyces cerevisiae. Biotechnol Lett 2021; 43:1735-1745. [PMID: 34047865 DOI: 10.1007/s10529-021-03151-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/25/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To study the effect of the mutation in conserved G412E in Cct7p subunit of CCT complex on its cellular fate. RESULTS TriC/CCT is a dynamic multimeric protein that assists in protein folding in an energy-dependent manner. A point mutation in the ATP binding pocket in the equatorial domain of the Cct7p subunit delays the doubling time. The cell size was twice the wild type, and the formation of protein aggregates suggests disturbed folding of the proteins. Upon growing in stressful conditions of arsenous acid and cadmium chloride, the mutant was lethal in As3+ but grew well in Cd2+ with 10.5 µg cadmium uptake mg-1 compared to the wild type. The increased expression of vacuole transporters YCF1 and BPT1 by ten-fold and two-fold in mutant indicates the metal transportation to the vacuole. CONCLUSION CCT complex was vulnerable to the mutation in G412E in the Cct7p subunit of protein folding molecular machinery. Interestingly, already stressed cells provided robustness against oxidative stress and cadmium sequestration in the vacuole.
Collapse
|
11
|
Abstract
Human papillomavirus (HPV) infection is a multi-step process that implies complex interactions of the viral particles with cellular proteins. The HPV capsid includes the two structural proteins L1 and L2, that play crucial roles on infectious viral entry. L2 is particularly relevant for the intracellular trafficking of the viral DNA towards the nucleus. Here, using proteomic studies we identified CCT proteins as novel interaction partners of HPV-16 L2. The CCT multimeric complex is an essential chaperonin which interacts with a large number of protein targets. We analysed the binding of different components of the CCT complex to L2. We confirmed the interaction of this structural viral protein with the CCT subunit 3 (CCT3) and we found that this interaction requires the N-terminal region of L2. Defects in HPV-16 pseudoviral particle (PsVs) infection were revealed by siRNA-mediated knockdown of some CCT subunits. While a substantial drop in the viral infection was associated with the ablation of CCT component 2, even more pronounced effects on infectivity were observed upon depletion of CCT component 3. Using confocal immunofluorescence assays, CCT3 co-localised with HPV PsVs at early times after infection, with L2 being required for this to occur. Further analysis showed the colocalization of several other subunits of CCT with the PsVs. Moreover, we observed a defect in capsid uncoating and a change in PsVs intracellular normal processing when ablating CCT3. Taken together, these studies demonstrate the importance of CCT chaperonin during HPV infectious entry.ImportanceSeveral of the mechanisms that function during the infection of target cells by HPV particles have been previously described. However, many aspects of this process remain unknown. In particular, the role of cellular proteins functioning as molecular chaperones during HPV infections has been only partially investigated. To the best of our knowledge, we describe here for the first time, a requirement of the CCT chaperonin for HPV infection. The role of this cellular complex seems to be determined by the binding of its component 3 to the viral structural protein L2. However, CCT's effect on HPV infection most probably comprises the whole chaperonin complex. Altogether, these studies define an important role for the CCT chaperonin in the processing and intracellular trafficking of HPV particles and in subsequent viral infectious entry.
Collapse
|
12
|
Zhang Y, Krieger J, Mikulska-Ruminska K, Kaynak B, Sorzano COS, Carazo JM, Xing J, Bahar I. State-dependent sequential allostery exhibited by chaperonin TRiC/CCT revealed by network analysis of Cryo-EM maps. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 160:104-120. [PMID: 32866476 PMCID: PMC7914283 DOI: 10.1016/j.pbiomolbio.2020.08.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 06/25/2020] [Accepted: 08/16/2020] [Indexed: 12/17/2022]
Abstract
The eukaryotic chaperonin TRiC/CCT plays a major role in assisting the folding of many proteins through an ATP-driven allosteric cycle. Recent structures elucidated by cryo-electron microscopy provide a broad view of the conformations visited at various stages of the chaperonin cycle, including a sequential activation of its subunits in response to nucleotide binding. But we lack a thorough mechanistic understanding of the structure-based dynamics and communication properties that underlie the TRiC/CCT machinery. In this study, we present a computational methodology based on elastic network models adapted to cryo-EM density maps to gain a deeper understanding of the structure-encoded allosteric dynamics of this hexadecameric machine. We have analysed several structures of the chaperonin resolved in different states toward mapping its conformational landscape. Our study indicates that the overall architecture intrinsically favours cooperative movements that comply with the structural variabilities observed in experiments. Furthermore, the individual subunits CCT1-CCT8 exhibit state-dependent sequential events at different states of the allosteric cycle. For example, in the ATP-bound state, subunits CCT5 and CCT4 selectively initiate the lid closure motions favoured by the overall architecture; whereas in the apo form of the heteromer, the subunit CCT7 exhibits the highest predisposition to structural change. The changes then propagate through parallel fluxes of allosteric signals to neighbours on both rings. The predicted state-dependent mechanisms of sequential activation provide new insights into TRiC/CCT intra- and inter-ring signal transduction events.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Computational and Systems Biology, University of Pittsburgh, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, PA, 15261, USA
| | - James Krieger
- Department of Computational and Systems Biology, University of Pittsburgh, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, PA, 15261, USA
| | - Karolina Mikulska-Ruminska
- Department of Computational and Systems Biology, University of Pittsburgh, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, PA, 15261, USA
| | - Burak Kaynak
- Department of Computational and Systems Biology, University of Pittsburgh, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, PA, 15261, USA
| | | | - José-María Carazo
- Centro Nacional de Biotecnología (CSIC), Darwin, 3, 28049, Madrid, Spain
| | - Jianhua Xing
- Department of Computational and Systems Biology, University of Pittsburgh, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, PA, 15261, USA
| | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
13
|
Temiz E, Koyuncu İ, Sahin E. CCT3 suppression prompts apoptotic machinery through oxidative stress and energy deprivation in breast and prostate cancers. Free Radic Biol Med 2021; 165:88-99. [PMID: 33508424 DOI: 10.1016/j.freeradbiomed.2021.01.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/20/2022]
Abstract
Mediated by chaperon proteins, protein misfolding plays a crucial role in cancer pathogenesis. Chaperonin Containing TCP1 Subunit 3 (CCT3) is one of eight subunits forming eukaryotic chaperons that catalyzes correct folding of the proteins employed in cell division, proliferation, and apoptosis pathway. Moreover, CCT3 expression increases responsively with carcinogenesis. However, how CCT3 drives the cancerous process has not been documented. Here we probed the mechanistic and functional interactions between CCT3 and apoptotic pathways and cell stressors. First, we profiled CCT3 expression levels of different 16 cell lines and found that CCT3 expression levels of CRL-2329 and PC3 were significantly increased. Then, we suppressed CCT3 levels in CRL-2329 and PC3 lines by miR-24-3p, miR-128-3p, and miR-149-5p mimics, and measured apoptotic response of the cell lines to the knockdown of CCT3 by acridine orange/ethidium bromide and Annexin V/PI staining, cell-cycle and mitochondria membrane potential (MMP) analyses, intracellular reactive oxygen species (ROS) measurement and analysis of expression levels of the apoptotic genes. After having suppressed CCT3, the cell cycle was arrested in the G0/G1 phase, MMP was impaired, and the intracellular ROS level was increased. These signs of apoptotic flux were corroborated by morphological images, statistically enhanced expression levels of the apoptotic pathway modulators and intracellular free amino acids profile. The free amino acid profile, which is heavily implicated in energy metabolism and cell division, is fluctuated in the progress of canceration. Strikingly, suppressed CCT3 shifted intracellular levels of glutamine, beta-alanine, glycine, serin, asparagine and sarcosine, which are employed in energy metabolism. Consequently, miRNA-mediated CCT3 suppression spur apoptosis by unbalancing the homeostasis in intracellular ROS and the profile of free amino acids in energy metabolism. Taken together, we anticipate that miRNA-mediated CCT3 suppression might provide a "dual therapeutic strategy" through conventional cellular toxicity as well as energy withdrawal.
Collapse
Affiliation(s)
- Ebru Temiz
- Program of Medical Promotion and Marketing, Health Services Vocational School, Harran University, Sanliurfa, Turkey; Department of Medical Biology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - İsmail Koyuncu
- Department of Medical Biochemistry, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - Emel Sahin
- Department of Immunology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey.
| |
Collapse
|
14
|
Fusella F, Seclì L, Cannata C, Brancaccio M. The one thousand and one chaperones of the NF-κB pathway. Cell Mol Life Sci 2020; 77:2275-2288. [PMID: 31811308 PMCID: PMC11104964 DOI: 10.1007/s00018-019-03402-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/04/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022]
Abstract
The NF-κB pathway represents a crucial signaling mechanism in sensing and integrating a multitude of environmental and intracellular stimuli and directing a coordinated response that from the cellular level may impact on the entire organism. A plethora of chaperone proteins work at multiple steps of the pathway, from membrane receptor activation to transcription factor binding to DNA. Indeed, chaperones are required to assist protein conformational changes, to assemble supramolecular complexes and to regulate protein ubiquitination, required for pathway activation. Some chaperones acquired a role as integral components of the signaling complexes, needed for signal progression. Here we describe the chaperones involved in the NF-κB pathway and their specific roles in the different contexts.
Collapse
Affiliation(s)
- Federica Fusella
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Laura Seclì
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Cristiana Cannata
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.
| |
Collapse
|
15
|
TRiC/CCT Complex, a Binding Partner of NS1 Protein, Supports the Replication of Zika Virus in Both Mammalians and Mosquitoes. Viruses 2020; 12:v12050519. [PMID: 32397176 PMCID: PMC7290343 DOI: 10.3390/v12050519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/02/2020] [Accepted: 05/06/2020] [Indexed: 11/23/2022] Open
Abstract
Mosquito-borne Zika virus (ZIKV) can cause congenital microcephaly and Guillain–Barré syndrome, among other symptoms. Specific treatments and vaccines for ZIKV are not currently available. To further understand the host factors that support ZIKV replication, we used mass spectrometry to characterize mammalian proteins that associate with the ZIKV NS1 protein and identified the TRiC/CCT complex as an interacting partner. Furthermore, the suppression of CCT2, one of the critical components of the TRiC/CCT complex, inhibited ZIKV replication in both mammalian cells and mosquitoes. These results highlight an important role for the TRiC/CCT complex in ZIKV infection, suggesting that the TRiC/CCT complex may be a promising therapeutic target.
Collapse
|
16
|
Galinato M, Shimoda K, Aguiar A, Hennig F, Boffelli D, McVoy MA, Hertel L. Single-Cell Transcriptome Analysis of CD34 + Stem Cell-Derived Myeloid Cells Infected With Human Cytomegalovirus. Front Microbiol 2019; 10:577. [PMID: 30949159 PMCID: PMC6437045 DOI: 10.3389/fmicb.2019.00577] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/06/2019] [Indexed: 12/18/2022] Open
Abstract
Myeloid cells are important sites of lytic and latent infection by human cytomegalovirus (CMV). We previously showed that only a small subset of myeloid cells differentiated from CD34+ hematopoietic stem cells is permissive to CMV replication, underscoring the heterogeneous nature of these populations. The exact identity of resistant and permissive cell types, and the cellular features characterizing the latter, however, could not be dissected using averaging transcriptional analysis tools such as microarrays and, hence, remained enigmatic. Here, we profile the transcriptomes of ∼7000 individual cells at day 1 post-infection using the 10× genomics platform. We show that viral transcripts are detectable in the majority of the cells, suggesting that virion entry is unlikely to be the main target of cellular restriction mechanisms. We further show that viral replication occurs in a small but specific sub-group of cells transcriptionally related to, and likely derived from, a cluster of cells expressing markers of Colony Forming Unit – Granulocyte, Erythrocyte, Monocyte, Megakaryocyte (CFU-GEMM) oligopotent progenitors. Compared to the remainder of the population, CFU-GEMM cells are enriched in transcripts with functions in mitochondrial energy production, cell proliferation, RNA processing and protein synthesis, and express similar or higher levels of interferon-related genes. While expression levels of the former are maintained in infected cells, the latter are strongly down-regulated. We thus propose that the preferential infection of CFU-GEMM cells may be due to the presence of a pre-established pro-viral environment, requiring minimal optimization efforts from viral effectors, rather than to the absence of specific restriction factors. Together, these findings identify a potentially new population of myeloid cells permissive to CMV replication, and provide a possible rationale for their preferential infection.
Collapse
Affiliation(s)
- Melissa Galinato
- Center for Immunobiology and Vaccine Development, Children's Hospital Oakland Research Institute, Oakland, CA, United States
| | - Kristen Shimoda
- Center for Immunobiology and Vaccine Development, Children's Hospital Oakland Research Institute, Oakland, CA, United States
| | - Alexis Aguiar
- Center for Immunobiology and Vaccine Development, Children's Hospital Oakland Research Institute, Oakland, CA, United States
| | - Fiona Hennig
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, United States
| | - Dario Boffelli
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, United States
| | - Michael A McVoy
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA, United States
| | - Laura Hertel
- Center for Immunobiology and Vaccine Development, Children's Hospital Oakland Research Institute, Oakland, CA, United States
| |
Collapse
|
17
|
The structure and evolution of eukaryotic chaperonin-containing TCP-1 and its mechanism that folds actin into a protein spring. Biochem J 2018; 475:3009-3034. [DOI: 10.1042/bcj20170378] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/16/2018] [Accepted: 08/28/2018] [Indexed: 12/15/2022]
Abstract
Actin is folded to its native state in eukaryotic cytosol by the sequential allosteric mechanism of the chaperonin-containing TCP-1 (CCT). The CCT machine is a double-ring ATPase built from eight related subunits, CCT1–CCT8. Non-native actin interacts with specific subunits and is annealed slowly through sequential binding and hydrolysis of ATP around and across the ring system. CCT releases a folded but soft ATP-G-actin monomer which is trapped 80 kJ/mol uphill on the folding energy surface by its ATP-Mg2+/Ca2+ clasp. The energy landscape can be re-explored in the actin filament, F-actin, because ATP hydrolysis produces dehydrated and more compact ADP-actin monomers which, upon application of force and strain, are opened and closed like the elements of a spring. Actin-based myosin motor systems underpin a multitude of force generation processes in cells and muscles. We propose that the water surface of F-actin acts as a low-binding energy, directional waveguide which is recognized specifically by the myosin lever-arm domain before the system engages to form the tight-binding actomyosin complex. Such a water-mediated recognition process between actin and myosin would enable symmetry breaking through fast, low energy initial binding events. The origin of chaperonins and the subsequent emergence of the CCT–actin system in LECA (last eukaryotic common ancestor) point to the critical role of CCT in facilitating phagocytosis during early eukaryotic evolution and the transition from the bacterial world. The coupling of CCT-folding fluxes to the cell cycle, cell size control networks and cancer are discussed together with directions for further research.
Collapse
|
18
|
Pouchucq L, Lobos-Ruiz P, Araya G, Valpuesta JM, Monasterio O. The chaperonin CCT promotes the formation of fibrillar aggregates of γ-tubulin. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1866:519-526. [PMID: 29339327 DOI: 10.1016/j.bbapap.2018.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 12/29/2022]
Abstract
The type II chaperonin CCT is involved in the prevention of the pathogenesis of numerous human misfolding disorders, as it sequesters misfolded proteins, blocks their aggregation and helps them to achieve their native state. In addition, it has been reported that CCT can prevent the toxicity of non-client amyloidogenic proteins by the induction of non-toxic aggregates, leading to new insight in chaperonin function as an aggregate remodeling factor. Here we add experimental evidence to this alternative mechanism by which CCT actively promotes the formation of conformationally different aggregates of γ-tubulin, a non-amyloidogenic CCT client protein, which are mediated by specific CCT-γ-tubulin interactions. The in vitro-induced aggregates were in some cases long fiber polymers, which compete with the amorphous aggregates. Direct injection of unfolded purified γ-tubulin into single-cell zebra fish embryos allowed us to relate this in vitro activity with the in vivo formation of intracellular aggregates. Injection of a CCT-binding deficient γ-tubulin mutant dramatically diminished the size of the intracellular aggregates, increasing the toxicity of the misfolded protein. These results point to CCT having a role in the remodeling of aggregates, constituting one of its many functions in cellular proteostasis.
Collapse
Affiliation(s)
- Luis Pouchucq
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile; Laboratorio de Biotecnología Vegetal Ambiental, Departamento de Biotecnología, Universidad Tecnológica Metropolitana, Santiago, Chile
| | - Pablo Lobos-Ruiz
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Gissela Araya
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - José María Valpuesta
- Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología (CNB-CSIC), Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Octavio Monasterio
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
19
|
Mazumdar R, Endler L, Monoyios A, Hess M, Bilic I. Establishment of a de novo Reference Transcriptome of Histomonas meleagridis Reveals Basic Insights About Biological Functions and Potential Pathogenic Mechanisms of the Parasite. Protist 2017; 168:663-685. [DOI: 10.1016/j.protis.2017.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/21/2017] [Accepted: 09/23/2017] [Indexed: 12/28/2022]
|
20
|
Melkani GC, Bhide S, Han A, Vyas J, Livelo C, Bodmer R, Bernstein SI. TRiC/CCT chaperonins are essential for maintaining myofibril organization, cardiac physiological rhythm, and lifespan. FEBS Lett 2017; 591:3447-3458. [PMID: 28963798 PMCID: PMC5683924 DOI: 10.1002/1873-3468.12860] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/08/2017] [Accepted: 09/19/2017] [Indexed: 01/12/2023]
Abstract
We recently reported that CCT chaperonin subunits are upregulated in a cardiac-specific manner under time-restricted feeding (TRF) [Gill S et al. (2015) Science 347, 1265-1269], suggesting that TRiC/CCT has a heart-specific function. To understand the CCT chaperonin function in cardiomyocytes, we performed its cardiac-specific knock-down in the Drosophila melanogaster model. This resulted in disorganization of cardiac actin- and myosin-containing myofibrils and severe physiological dysfunction, including restricted heart diameters, elevated cardiac dysrhythmia and compromised cardiac performance. We also noted that cardiac-specific knock-down of CCT chaperonin significantly shortens lifespans. Additionally, disruption of circadian rhythm yields further deterioration of cardiac function of hypomorphic CCT mutants. Our analysis reveals that both the orchestration of protein folding and circadian rhythms mediated by CCT chaperonin are critical for maintaining heart contractility.
Collapse
Affiliation(s)
- Girish C. Melkani
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| | - Shruti Bhide
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| | - Andrew Han
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| | - Jay Vyas
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| | - Catherine Livelo
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sanford I. Bernstein
- Department of Biology, Molecular Biology and Heart Institutes, San Diego State University San Diego, CA 92182, USA
| |
Collapse
|
21
|
Di Genova BM, da Silva RC, da Cunha JPC, Gargantini PR, Mortara RA, Tonelli RR. Protein SUMOylation is Involved in Cell-cycle Progression and Cell Morphology in Giardia lamblia. J Eukaryot Microbiol 2016; 64:491-503. [PMID: 27864857 DOI: 10.1111/jeu.12386] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 10/13/2016] [Accepted: 11/02/2016] [Indexed: 01/08/2023]
Abstract
The unicellular protozoa Giardia lamblia is a food- and waterborne parasite that causes giardiasis. This illness is manifested as acute and self-limited diarrhea and can evolve to long-term complications. Successful establishment of infection by Giardia trophozoites requires adhesion to host cells and colonization of the small intestine, where parasites multiply by mitotic division. The tight binding of trophozoites to host cells occurs by means of the ventral adhesive disc, a spiral array of microtubules and associated proteins such as giardins. In this work we show that knock down of the Small Ubiquitin-like MOdifier (SUMO) results in less adhesive trophzoites, decreased cell proliferation and deep morphological alterations, including at the ventral disc. Consistent with the reduced proliferation, SUMO knocked-down trophozoites were arrested in G1 and in S phases of the cell cycle. Mass spectrometry analysis of anti-SUMO immunoprecipitates was performed to identify SUMO substrates possibly involved in these events. Among the identified SUMOylation targets, α-tubulin was further validated by Western blot and confirmed to be a SUMO target in Giardia trophozoites.
Collapse
Affiliation(s)
- Bruno M Di Genova
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, RuaBotucatu 862, 04023-062, Vila Clementino, São Paulo, SP, Brazil
| | - Richard C da Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, RuaBotucatu 862, 04023-062, Vila Clementino, São Paulo, SP, Brazil
| | - Júlia P C da Cunha
- Laboratório Especial de Ciclo Celular, Centro de Toxinas, Resposta Imune e Sinalização Celular - CeTICS, Instituto Butantan, Avenida Vital Brasil 1500, 05503-900, Butantã, São Paulo, SP, Brazil
| | - Pablo R Gargantini
- Laboratorio de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Católica de Córdoba (UCC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Armada Argentina 3555., X5016DHK, Cordoba, Argentina
| | - Renato A Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, RuaBotucatu 862, 04023-062, Vila Clementino, São Paulo, SP, Brazil
| | - Renata R Tonelli
- Departamento de Ciências Biológicas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Rua São Nicolau 210, 09913-030, Diadema, SP, Brazil
| |
Collapse
|
22
|
VanPelt J, Page RC. Unraveling the CHIP:Hsp70 complex as an information processor for protein quality control. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1865:133-141. [PMID: 27863257 DOI: 10.1016/j.bbapap.2016.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/11/2016] [Accepted: 11/14/2016] [Indexed: 11/24/2022]
Abstract
The CHIP:Hsp70 complex stands at the crossroads of the cellular protein quality control system. Hsp70 facilitates active refolding of misfolded client proteins, while CHIP directs ubiquitination of misfolded client proteins bound to Hsp70. The direct competition between CHIP and Hsp70 for the fate of misfolded proteins leads to the question: how does the CHIP:Hsp70 complex execute triage decisions that direct misfolded proteins for either refolding or degradation? The current body of literature points toward action of the CHIP:Hsp70 complex as an information processor that takes inputs in the form of client folding state, dynamics, and posttranslational modifications, then outputs either refolded or ubiquitinated client proteins. Herein we examine the CHIP:Hsp70 complex beginning with the structure and function of CHIP and Hsp70, followed by an examination of recent studies of the interactions and dynamics of the CHIP:Hsp70 complex.
Collapse
Affiliation(s)
- Jamie VanPelt
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA
| | - Richard C Page
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
23
|
Génier S, Degrandmaison J, Moreau P, Labrecque P, Hébert TE, Parent JL. Regulation of GPCR expression through an interaction with CCT7, a subunit of the CCT/TRiC complex. Mol Biol Cell 2016; 27:3800-3812. [PMID: 27708139 PMCID: PMC5170604 DOI: 10.1091/mbc.e16-04-0224] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 09/22/2016] [Accepted: 09/26/2016] [Indexed: 12/25/2022] Open
Abstract
A direct and functional interaction between a subunit of the CCT/TCP-1 ring complex (TRiC) chaperonin complex and G protein–coupled receptor (GPCRs) is shown. Evidence is provided that distinct nascent GPCRs can undergo alternative folding pathways and that CCT/TRiC is critical in preventing aggregation of some GPCRs and in promoting their proper maturation and expression. Mechanisms that prevent aggregation and promote folding of nascent G protein–coupled receptors (GPCRs) remain poorly understood. We identified chaperonin containing TCP-1 subunit eta (CCT7) as an interacting partner of the β-isoform of thromboxane A2 receptor (TPβ) by yeast two-hybrid screening. CCT7 coimmunoprecipitated with overexpressed TPβ and β2-adrenergic receptor (β2AR) in HEK 293 cells, but also with endogenous β2AR. CCT7 depletion by small interfering RNA reduced total and cell-surface expression of both receptors and caused redistribution of the receptors to juxtanuclear aggresomes, significantly more so for TPβ than β2AR. Interestingly, Hsp90 coimmunoprecipitated with β2AR but virtually not with TPβ, indicating that nascent GPCRs can adopt alternative folding pathways. In vitro pull-down assays showed that both receptors can interact directly with CCT7 through their third intracellular loops and C-termini. We demonstrate that Trp334 in the TPβ C-terminus is critical for the CCT7 interaction and plays an important role in TPβ maturation and cell-surface expression. Of note, introducing a tryptophan in the corresponding position of the TPα isoform confers the CCT7-binding and maturation properties of TPβ. We show that an interaction with a subunit of the CCT/TCP-1 ring complex (TRiC) chaperonin complex is involved in regulating aggregation of nascent GPCRs and in promoting their proper maturation and expression.
Collapse
Affiliation(s)
- Samuel Génier
- Service de Rhumatologie, Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CR-CHUS), and Institut de Pharmacologie de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jade Degrandmaison
- Service de Rhumatologie, Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CR-CHUS), and Institut de Pharmacologie de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Pierrick Moreau
- Service de Rhumatologie, Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CR-CHUS), and Institut de Pharmacologie de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Pascale Labrecque
- Service de Rhumatologie, Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CR-CHUS), and Institut de Pharmacologie de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Jean-Luc Parent
- Service de Rhumatologie, Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CR-CHUS), and Institut de Pharmacologie de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
24
|
TRiC subunits enhance BDNF axonal transport and rescue striatal atrophy in Huntington's disease. Proc Natl Acad Sci U S A 2016; 113:E5655-64. [PMID: 27601642 DOI: 10.1073/pnas.1603020113] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Corticostriatal atrophy is a cardinal manifestation of Huntington's disease (HD). However, the mechanism(s) by which mutant huntingtin (mHTT) protein contributes to the degeneration of the corticostriatal circuit is not well understood. We recreated the corticostriatal circuit in microfluidic chambers, pairing cortical and striatal neurons from the BACHD model of HD and its WT control. There were reduced synaptic connectivity and atrophy of striatal neurons in cultures in which BACHD cortical and striatal neurons were paired. However, these changes were prevented if WT cortical neurons were paired with BACHD striatal neurons; synthesis and release of brain-derived neurotrophic factor (BDNF) from WT cortical axons were responsible. Consistent with these findings, there was a marked reduction in anterograde transport of BDNF in BACHD cortical neurons. Subunits of the cytosolic chaperonin T-complex 1 (TCP-1) ring complex (TRiC or CCT for chaperonin containing TCP-1) have been shown to reduce mHTT levels. Both CCT3 and the apical domain of CCT1 (ApiCCT1) decreased the level of mHTT in BACHD cortical neurons. In cortical axons, they normalized anterograde BDNF transport, restored retrograde BDNF transport, and normalized lysosomal transport. Importantly, treating BACHD cortical neurons with ApiCCT1 prevented BACHD striatal neuronal atrophy by enhancing release of BDNF that subsequently acts through tyrosine receptor kinase B (TrkB) receptor on striatal neurons. Our findings are evidence that TRiC reagent-mediated reductions in mHTT enhanced BDNF delivery to restore the trophic status of BACHD striatal neurons.
Collapse
|
25
|
Qian EN, Han SY, Ding SZ, Lv X. Expression and diagnostic value of CCT3 and IQGAP3 in hepatocellular carcinoma. Cancer Cell Int 2016; 16:55. [PMID: 27390551 PMCID: PMC4936258 DOI: 10.1186/s12935-016-0332-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/23/2016] [Indexed: 12/14/2022] Open
Abstract
Background To evaluate plasma chaperonin containing TCP1 complex subunit 3 (CCT3) and IQ-motif-containing GTPase-activating protein-3 (IQGAP3) as biomarker for hepatocellular carcinoma (HCC) screening and diagnosis. Methods Blood samples were collected from 126 HCC patients with HCC, 88 patients with cirrhosis and 50 healthy volunteers to detect plasma α-fetoprotein (AFP), CCT3 and IQGAP3 levels. Plasma AFP, CCT3 and IQGAP3 protein levels were measured by enzyme linked immunosorbent assay (ELISA). Results In the plasma of HCC patients, both CCT3 and IQGAP3 were significantly higher than in patients with cirrhosis and in healthy controls (P < 0.01). CCT3 and IQGAP3 protein level correlated well with HCC etiology, tumor size, TNM stage, and child-pugh classification. CCT3 protein had higher sensitivity in the diagnosis of HCC when compared with AFP (87.3 vs 69.8 %). In addition, CCT3 and IQGAP3 protein were able to complement AFP in detecting AFP-negative HCC patients with sensitivity and specificity of 92.1 and 70.5 % and 81.6 and 71.6 %, respectively. In the small HCC group, CCT3 and IQGAP3 protein had sensitivity of 76.6 and 74.5 %, respectively. The combination of AFP + CCT3 + IQGAP3 (0.954) had significantly superior discriminative ability than AFP alone (0.815; P < 0.01). Conclusions CCT3 and IQGAP3 are novel complementary biomarkers for HCC screening and diagnosis, especially for AFP-negative and small HCC patients.
Collapse
Affiliation(s)
- E-Na Qian
- Department of Gastroenterology and Hepatology, People's Hospital of Zhengzhou University, No. 7 Wei Wu Road, Zhengzhou, 450003 Henan China
| | - Shuang-Yin Han
- Department of Gastroenterology and Hepatology, People's Hospital of Zhengzhou University, No. 7 Wei Wu Road, Zhengzhou, 450003 Henan China
| | - Song-Ze Ding
- Department of Gastroenterology and Hepatology, People's Hospital of Zhengzhou University, No. 7 Wei Wu Road, Zhengzhou, 450003 Henan China
| | - Xun Lv
- Department of Gastroenterology and Hepatology, People's Hospital of Zhengzhou University, No. 7 Wei Wu Road, Zhengzhou, 450003 Henan China
| |
Collapse
|
26
|
Blanc M, David F, Abrami L, Migliozzi D, Armand F, Bürgi J, van der Goot FG. SwissPalm: Protein Palmitoylation database. F1000Res 2015; 4:261. [PMID: 26339475 PMCID: PMC4544385 DOI: 10.12688/f1000research.6464.1] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2015] [Indexed: 12/19/2022] Open
Abstract
Protein S-palmitoylation is a reversible post-translational modification that regulates many key biological processes, although the full extent and functions of protein S-palmitoylation remain largely unexplored. Recent developments of new chemical methods have allowed the establishment of palmitoyl-proteomes of a variety of cell lines and tissues from different species. As the amount of information generated by these high-throughput studies is increasing, the field requires centralization and comparison of this information. Here we present SwissPalm (
http://swisspalm.epfl.ch), our open, comprehensive, manually curated resource to study protein S-palmitoylation. It currently encompasses more than 5000 S-palmitoylated protein hits from seven species, and contains more than 500 specific sites of S-palmitoylation. SwissPalm also provides curated information and filters that increase the confidence in true positive hits, and integrates predictions of S-palmitoylated cysteine scores, orthologs and isoform multiple alignments. Systems analysis of the palmitoyl-proteome screens indicate that 10% or more of the human proteome is susceptible to S-palmitoylation. Moreover, ontology and pathway analyses of the human palmitoyl-proteome reveal that key biological functions involve this reversible lipid modification. Comparative analysis finally shows a strong crosstalk between S-palmitoylation and other post-translational modifications. Through the compilation of data and continuous updates, SwissPalm will provide a powerful tool to unravel the global importance of protein S-palmitoylation.
Collapse
Affiliation(s)
- Mathieu Blanc
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Fabrice David
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland.,Bioinformatics and biostatistics Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Laurence Abrami
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Daniel Migliozzi
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Florence Armand
- Proteomic Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Jérôme Bürgi
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Françoise Gisou van der Goot
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| |
Collapse
|
27
|
iTRAQ-based quantitative proteomics study on the neuroprotective effects of extract of Acanthopanax senticosus harm on SH-SY5Y cells overexpressing A53T mutant α-synuclein. Neurochem Int 2014; 72:37-47. [DOI: 10.1016/j.neuint.2014.04.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/04/2014] [Accepted: 04/08/2014] [Indexed: 01/10/2023]
|
28
|
Kypri E, Christodoulou A, Maimaris G, Lethan M, Markaki M, Lysandrou C, Lederer CW, Tavernarakis N, Geimer S, Pedersen LB, Santama N. The nucleotide-binding proteins Nubp1 and Nubp2 are negative regulators of ciliogenesis. Cell Mol Life Sci 2014; 71:517-38. [PMID: 23807208 PMCID: PMC11113914 DOI: 10.1007/s00018-013-1401-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 06/03/2013] [Accepted: 06/06/2013] [Indexed: 12/12/2022]
Abstract
Nucleotide-binding proteins Nubp1 and Nubp2 are MRP/MinD-type P-loop NTPases with sequence similarity to bacterial division site-determining proteins and are conserved, essential proteins throughout the Eukaryotes. They have been implicated, together with their interacting minus-end directed motor protein KIFC5A, in the regulation of centriole duplication in mammalian cells. Here we show that Nubp1 and Nubp2 are integral components of centrioles throughout the cell cycle, recruited independently of KIFC5A. We further demonstrate their localization at the basal body of the primary cilium in quiescent vertebrate cells or invertebrate sensory cilia, as well as in the motile cilia of mouse cells and in the flagella of Chlamydomonas. RNAi-mediated silencing of nubp-1 in C. elegans causes the formation of morphologically aberrant and additional cilia in sensory neurons. Correspondingly, downregulation of Nubp1 or Nubp2 in mouse quiescent NIH 3T3 cells markedly increases the number of ciliated cells, while knockdown of KIFC5A dramatically reduces ciliogenesis. Simultaneous double silencing of Nubp1 + KIFC5A restores the percentage of ciliated cells to control levels. We document the normal ciliary recruitment, during these silencing regimes, of basal body proteins critical for ciliogenesis, namely CP110, CEP290, cenexin, Chibby, AurA, Rab8, and BBS7. Interestingly, we uncover novel interactions of Nubp1 with several members of the CCT/TRiC molecular chaperone complex, which we find enriched at the basal body and recruited independently of the Nubps or KIFC5A. Our combined results for Nubp1, Nubp2, and KIFC5A and their striking effects on cilium formation suggest a central regulatory role for these proteins, likely involving CCT/TRiC chaperone activity, in ciliogenesis.
Collapse
Affiliation(s)
- Elena Kypri
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| | - Andri Christodoulou
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| | - Giannis Maimaris
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| | - Mette Lethan
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Maria Markaki
- Institute of Molecular Biology and Biotechnology, Crete, Greece
| | - Costas Lysandrou
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| | | | | | | | - Lotte B. Pedersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Niovi Santama
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| |
Collapse
|
29
|
Na I, Reddy KD, Breydo L, Xue B, Uversky VN. A putative role of the Sup35p C-terminal domain in the cytoskeleton organization during yeast mitosis. ACTA ACUST UNITED AC 2014; 10:925-40. [DOI: 10.1039/c3mb70515c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Based on structural analysis of several effectors and partners, Sup35pC is proposed to serve as actin modulator during mitosis.
Collapse
Affiliation(s)
- Insung Na
- Department of Molecular Medicine
- Morsani College of Medicine
- University of South Florida
- Tampa, USA
| | - Krishna D. Reddy
- Department of Molecular Medicine
- Morsani College of Medicine
- University of South Florida
- Tampa, USA
| | - Leonid Breydo
- Department of Molecular Medicine
- Morsani College of Medicine
- University of South Florida
- Tampa, USA
| | - Bin Xue
- Department of Cell Biology
- Microbiology, and Molecular Biology
- College of Arts and Science
- University of South Florida
- Tampa, USA
| | - Vladimir N. Uversky
- Department of Molecular Medicine
- Morsani College of Medicine
- University of South Florida
- Tampa, USA
- USF Health Byrd Alzheimer's Research Institute
| |
Collapse
|
30
|
Proteasome overload is a common stress factor in multiple forms of inherited retinal degeneration. Proc Natl Acad Sci U S A 2013; 110:9986-91. [PMID: 23716657 DOI: 10.1073/pnas.1305521110] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Inherited retinal degenerations, caused by mutations in over 100 individual genes, affect approximately 2 million people worldwide. Many of the underlying mutations cause protein misfolding or mistargeting in affected photoreceptors. This places an increased burden on the protein folding and degradation machinery, which may trigger cell death. We analyzed how these cellular functions are affected in degenerating rods of the transducin γ-subunit (Gγ1) knockout mouse. These rods produce large amounts of transducin β-subunit (Gβ1), which cannot fold without Gγ1 and undergoes intracellular proteolysis instead of forming a transducin βγ-subunit complex. Our data revealed that the most critical pathobiological factor leading to photoreceptor cell death in these animals is insufficient capacity of proteasomes to process abnormally large amounts of misfolded protein. A decrease in the Gβ1 production in Gγ1 knockout rods resulted in a significant reduction in proteasomal overload and caused a striking reversal of photoreceptor degeneration. We further demonstrated that a similar proteasomal overload takes place in photoreceptors of other mutant mice where retinal degeneration has been ascribed to protein mistargeting or misfolding, but not in mice whose photoreceptor degenerate as a result of abnormal phototransduction. These results establish the prominence of proteasomal insufficiency across multiple degenerative diseases of the retina, thereby positioning proteasomes as a promising therapeutic target for treating these debilitating conditions.
Collapse
|
31
|
Cellular chaperonin CCTγ contributes to rabies virus replication during infection. J Virol 2013; 87:7608-21. [PMID: 23637400 DOI: 10.1128/jvi.03186-12] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Rabies, as the oldest known infectious disease, remains a serious threat to public health worldwide. The eukaryotic cytosolic chaperonin TRiC/CCT complex facilitates the folding of proteins through ATP hydrolysis. Here, we investigated the expression, cellular localization, and function of neuronal CCTγ during neurotropic rabies virus (RABV) infection using mouse N2a cells as a model. Following RABV infection, 24 altered proteins were identified by using two-dimensional electrophoresis and mass spectrometry, including 20 upregulated proteins and 4 downregulated proteins. In mouse N2a cells infected with RABV or cotransfected with RABV genes encoding nucleoprotein (N) and phosphoprotein (P), confocal microscopy demonstrated that upregulated cellular CCTγ was colocalized with viral proteins N and P, which formed a hollow cricoid inclusion within the region around the nucleus. These inclusions, which correspond to Negri bodies (NBs), did not form in mouse N2a cells only expressing the viral protein N or P. Knockdown of CCTγ by lentivirus-mediated RNA interference led to significant inhibition of RABV replication. These results demonstrate that the complex consisting of viral proteins N and P recruits CCTγ to NBs and identify the chaperonin CCTγ as a host factor that facilitates intracellular RABV replication. This work illustrates how viruses can utilize cellular chaperonins and compartmentalization for their own benefit.
Collapse
|
32
|
Knee KM, Sergeeva OA, King JA. Human TRiC complex purified from HeLa cells contains all eight CCT subunits and is active in vitro. Cell Stress Chaperones 2013; 18:137-44. [PMID: 23011926 PMCID: PMC3581623 DOI: 10.1007/s12192-012-0357-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 07/10/2012] [Accepted: 07/13/2012] [Indexed: 10/28/2022] Open
Abstract
Archaeal and eukaryotic cytosols contain group II chaperonins, which have a double-barrel structure and fold proteins inside a cavity in an ATP-dependent manner. The most complex of the chaperonins, the eukaryotic TCP-1 ring complex (TRiC), has eight different subunits, chaperone containing TCP-1 (CCT1-8), that are arranged so that there is one of each subunit per ring. Aspects of the structure and function of the bovine and yeast TRiC have been characterized, but studies of human TRiC have been limited. We have isolated and purified endogenous human TRiC from HeLa suspension cells. This purified human TRiC contained all eight CCT subunits organized into double-barrel rings, consistent with what has been found for bovine and yeast TRiC. The purified human TRiC is active as demonstrated by the luciferase refolding assay. As a more stringent test, the ability of human TRiC to suppress the aggregation of human γD-crystallin was examined. In addition to suppressing off-pathway aggregation, TRiC was able to assist the refolding of the crystallin molecules, an activity not found with the lens chaperone, α-crystallin. Additionally, we show that human TRiC from HeLa cell lysate is associated with the heat shock protein 70 and heat shock protein 90 chaperones. Purification of human endogenous TRiC from HeLa cells will enable further characterization of this key chaperonin, required for the reproduction of all human cells.
Collapse
Affiliation(s)
- Kelly M. Knee
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Ave., 68-330, Cambridge, MA 02139 USA
| | - Oksana A. Sergeeva
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Ave., 68-330, Cambridge, MA 02139 USA
| | - Jonathan A. King
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Ave., 68-330, Cambridge, MA 02139 USA
| |
Collapse
|