1
|
Castedo N, Alfonso A, Alvariño R, Vieytes MR, Botana LM. Cyclophilin A and C are the Main Components of Extracellular Vesicles in Response to Hyperglycemia in BV2 Microglial Cells. Mol Neurobiol 2025:10.1007/s12035-025-04921-6. [PMID: 40199808 DOI: 10.1007/s12035-025-04921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Cyclophilins (Cyps) and CD147 receptor play a crucial role in the inflammatory responses. Chronic inflammation causes tissue damage and is a common condition of several inflammation-based pathologies as diabetes or Alzheimer´s disease. Under high glucose (HG) conditions, microglia is activated and releases inflammatory mediators. In this process the role of Cyps is unknown, so this study was aimed to investigate the profile of Cyps in microglia and their release through extracellular vesicles (EVs) under hyperglycemia. An increase in reactive oxygen species (ROS) and nitric oxide (NO) levels was observed when BV2 glia cells were incubated with HG concentration. These effects were mitigated by the Cyps inhibitor cyclosporine A (CsA), suggesting the implication of Cyps in BV2 activation. In these conditions the intracellular expression of CypA, B, C and D, as well as the membrane expression of CD147 receptor was increased. In addition, only CypA and CypC were detected in the extracellular medium. Then, the presence of Cyps inside EVs was explored as an alternative secretion route. Interestingly, under HG treatment, an increase in the levels of the four Cyps in EVs was observed. When neurons were treated with EVs derived from HG-treated glia cells, their viability was reduced and EVs were detected in cytosol neurons pointing to an EVs-Cyps neurotoxic effect. These findings provide novel insights into the relationship between Cyps and EVs in neuroinflammation in hyperglycemia conditions. The current results strengthen the role of Cyps in cell communication and its potential role in brain function under pathological conditions.
Collapse
Affiliation(s)
- Noelia Castedo
- Departamento de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España
| | - Amparo Alfonso
- Departamento de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España
| | - Rebeca Alvariño
- Departamento de Fisiología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España.
| | - Mercedes R Vieytes
- Departamento de Fisiología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España
| | - Luis M Botana
- Departamento de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España.
| |
Collapse
|
2
|
Zhou B, Sha S, Wang Q, Sun S, Tao J, Zhu J, Dong L. The prognostic implications of cuproptosis-related gene signature and the potential of PPIC as a promising biomarker in cutaneous melanoma. Pigment Cell Melanoma Res 2024; 37:864-880. [PMID: 39115044 DOI: 10.1111/pcmr.13185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/28/2024] [Accepted: 06/23/2024] [Indexed: 10/29/2024]
Abstract
Cutaneous melanoma is the most lethal of all skin tumors. Recently, cuproptosis, a novel form of cell death linked to oxidative phosphorylation, has emerged as an important factor. However, the precise role of cuproptosis in melanoma remains unclear. Our research explored the potential links between cuproptosis-related genes, prognosis, immune microenvironments, and melanoma treatments. Significantly, cuproptosis regulators showed remarkable differences between melanoma and normal tissues, establishing their relevance to melanoma. The newly developed cuproptosis-related gene signature (CGS) demonstrated a robust ability to predict overall survival (OS) in melanoma. We constructed a novel nomogram that combined clinical features with CGS to improve predictive accuracy. In addition, the study revealed correlations between CGS and immune cell populations, including CD8+T cells, Tfh cells, B cells, and myeloid-derived suppressor cells. Within the CGS, Peptidylprolyl isomerase C (PPIC) emerged as the most strongly associated with poor prognosis and drug resistance in melanoma. PPIC was identified as a promoter of melanoma progression, enhancing cell invasiveness while concurrently suppressing CD8+T cell activation. This comprehensive study not only elucidated the intricate connections between CGS, melanoma prognosis, immune microenvironment, and drug resistance but also provided compelling evidence supporting PPIC as a promising biomarker for predicting OS in melanoma treatment.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Shanshan Sha
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Qi Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Shuomin Sun
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Juan Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Jinjin Zhu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| | - Liyun Dong
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, China
| |
Collapse
|
3
|
Tripp BA, Dillon ST, Yuan M, Asara JM, Vasunilashorn SM, Fong TG, Inouye SK, Ngo LH, Marcantonio ER, Xie Z, Libermann TA, Otu HH. Integrated Multi-Omics Analysis of Cerebrospinal Fluid in Postoperative Delirium. Biomolecules 2024; 14:924. [PMID: 39199312 PMCID: PMC11352186 DOI: 10.3390/biom14080924] [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: 06/29/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 09/01/2024] Open
Abstract
Preoperative risk biomarkers for delirium may aid in identifying high-risk patients and developing intervention therapies, which would minimize the health and economic burden of postoperative delirium. Previous studies have typically used single omics approaches to identify such biomarkers. Preoperative cerebrospinal fluid (CSF) from the Healthier Postoperative Recovery study of adults ≥ 63 years old undergoing elective major orthopedic surgery was used in a matched pair delirium case-no delirium control design. We performed metabolomics and lipidomics, which were combined with our previously reported proteomics results on the same samples. Differential expression, clustering, classification, and systems biology analyses were applied to individual and combined omics datasets. Probabilistic graph models were used to identify an integrated multi-omics interaction network, which included clusters of heterogeneous omics interactions among lipids, metabolites, and proteins. The combined multi-omics signature of 25 molecules attained an AUC of 0.96 [95% CI: 0.85-1.00], showing improvement over individual omics-based classification. We conclude that multi-omics integration of preoperative CSF identifies potential risk markers for delirium and generates new insights into the complex pathways associated with delirium. With future validation, this hypotheses-generating study may serve to build robust biomarkers for delirium and improve our understanding of its pathophysiology.
Collapse
Affiliation(s)
- Bridget A. Tripp
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Simon T. Dillon
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.)
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
| | - Min Yuan
- Division of Signal Transduction and Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - John M. Asara
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Division of Signal Transduction and Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Sarinnapha M. Vasunilashorn
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tamara G. Fong
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA 02131, USA
| | - Sharon K. Inouye
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA 02131, USA
| | - Long H. Ngo
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Edward R. Marcantonio
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Zhongcong Xie
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Towia A. Libermann
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.)
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Hasan H. Otu
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
4
|
Li J, Zhu J, Deng Y, Reck EC, Walker EM, Sidarala V, Hubers DL, Pasmooij MB, Shin CS, Bandesh K, Motakis E, Nargund S, Kursawe R, Basrur V, Nesvizhskii AI, Stitzel ML, Chan DC, Soleimanpour SA. LONP1 regulation of mitochondrial protein folding provides insight into beta cell failure in type 2 diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597215. [PMID: 38895283 PMCID: PMC11185607 DOI: 10.1101/2024.06.03.597215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Proteotoxicity is a contributor to the development of type 2 diabetes (T2D), but it is unknown whether protein misfolding in T2D is generalized or has special features. Here, we report a robust accumulation of misfolded proteins within the mitochondria of human pancreatic islets in T2D and elucidate its impact on β cell viability. Surprisingly, quantitative proteomics studies of protein aggregates reveal that human islets from donors with T2D have a signature more closely resembling mitochondrial rather than ER protein misfolding. The matrix protease LonP1 and its chaperone partner mtHSP70 were among the proteins enriched in protein aggregates. Deletion of LONP1 in mice yields mitochondrial protein misfolding and reduced respiratory function, ultimately leading to β cell apoptosis and hyperglycemia. Intriguingly, LONP1 gain of function ameliorates mitochondrial protein misfolding and restores human β cell survival following glucolipotoxicity via a protease-independent effect requiring LONP1-mtHSP70 chaperone activity. Thus, LONP1 promotes β cell survival and prevents hyperglycemia by facilitating mitochondrial protein folding. These observations may open novel insights into the nature of impaired proteostasis on β cell loss in the pathogenesis of T2D that could be considered as future therapeutic targets.
Collapse
|
5
|
Law ME, Dulloo ZM, Eggleston SR, Takacs GP, Alexandrow GM, Lee YI, Wang M, Hardy B, Su H, Forsyth B, Das P, Datta PK, Chiang CW, Sharma A, Kanumuri SRR, Guryanova OA, Harrison JK, Tirosh B, Castellano RK, Law BK. DR5 disulfide bonding functions as a sensor and effector of protein folding stress. Mol Cancer Res 2024:754250. [PMID: 40105733 PMCID: PMC11989202 DOI: 10.1158/1541-7786.mcr-24-0756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/17/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
New agents are needed that selectively kill cancer cells without harming normal tissues. The TRAIL ligand and its receptors, DR5 and DR4, exhibit cancer-selective toxicity. TRAIL analogs or agonistic antibodies targeting these receptors are available but have not yet received FDA approval for cancer therapy. Small molecules for activating DR5 or DR4 independently of protein ligands may activate TRAIL receptors as a monotherapy or potentiate the efficacy of TRAIL analogs and agonistic antibodies. Previously described Disulfide bond Disrupting Agents (DDAs) activate DR5 by altering its disulfide bonding through inhibition of the Protein Disulfide Isomerases (PDIs) ERp44, AGR2, and PDIA1. Work presented here extends these findings by showing that disruption of single DR5 disulfide bonds causes high-level DR5 expression, disulfide-mediated clustering, and activation of Caspase 8-Caspase 3 mediated pro-apoptotic signaling. Recognition of the extracellular domain of DR5 by various antibodies is strongly influenced by the pattern of DR5 disulfide bonding, which has important implications for the use of agonistic DR5 antibodies for cancer therapy and as research tools. Importantly, other ER stressors, including Thapsigargin and Tunicamycin also alter DR5 disulfide bonding in various cancer cell lines and in some instances, DR5 mis-disulfide bonding is potentiated by overriding the Integrated Stress Response (ISR) with inhibitors of the PERK kinase or the ISR inhibitor ISRIB. These observations indicate that the pattern of DR5 disulfide bonding functions as a sensor of ER stress and serves as an effector of proteotoxic stress by driving extrinsic apoptosis independently of extracellular ligands. Implications: Extreme endoplasmic reticulum stress triggers triage of transmembrane receptor production, whereby mitogenic receptors are downregulated and death receptors are simultaneously elevated.
Collapse
Affiliation(s)
- Mary E. Law
- Department of Pharmacology & Therapeutics, University of Florida, 32610
| | - Zaafir M. Dulloo
- Department of Chemistry, University of Florida, Gainesville, FL, 32611
| | | | - Gregory P. Takacs
- Department of Pharmacology & Therapeutics, University of Florida, 32610
| | | | - Young il Lee
- Department of Pharmacology & Therapeutics, University of Florida, 32610
| | - Mengxiong Wang
- Department of Radiation Biology, Stanford University, Stanford, CA, 94305
| | - Brian Hardy
- Department of Pharmacology & Therapeutics, University of Florida, 32610
| | - Hanyu Su
- Department of Pharmacology & Therapeutics, University of Florida, 32610
| | - Bianca Forsyth
- Department of Pharmacology & Therapeutics, University of Florida, 32610
| | - Parag Das
- Department of Chemistry, University of Florida, Gainesville, FL, 32611
| | - Pran K. Datta
- Division of Hematology & Oncology, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Chi-Wu Chiang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Abhisheak Sharma
- Department of Pharmaceutics, University of Florida, Gainesville, FL, 32610
| | | | - Olga A. Guryanova
- Department of Pharmacology & Therapeutics, University of Florida, 32610
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32610
| | - Jeffrey K. Harrison
- Department of Pharmacology & Therapeutics, University of Florida, 32610
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32610
| | - Boaz Tirosh
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106
| | - Ronald K. Castellano
- Department of Chemistry, University of Florida, Gainesville, FL, 32611
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32610
| | - Brian K. Law
- Department of Pharmacology & Therapeutics, University of Florida, 32610
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32610
| |
Collapse
|
6
|
Law ME, Dulloo ZM, Eggleston SR, Takacs GP, Alexandrow GM, Wang M, Su H, Forsyth B, Chiang CW, Sharma A, Kanumuri SRR, Guryanova OA, Harrison JK, Tirosh B, Castellano RK, Law BK. DR5 disulfide bonding as a sensor and effector of protein folding stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583390. [PMID: 38496520 PMCID: PMC10942403 DOI: 10.1101/2024.03.04.583390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
New agents are needed that selectively kill cancer cells without harming normal tissues. The TRAIL ligand and its receptors, DR5 and DR4, exhibit cancer-selective toxicity, but TRAIL analogs or agonistic antibodies targeting these receptors have not received FDA approval for cancer therapy. Small molecules for activating DR5 or DR4 independently of protein ligands may bypass some of the pharmacological limitations of these protein drugs. Previously described Disulfide bond Disrupting Agents (DDAs) activate DR5 by altering its disulfide bonding through inhibition of the Protein Disulfide Isomerases (PDIs) ERp44, AGR2, and PDIA1. Work presented here extends these findings by showing that disruption of single DR5 disulfide bonds causes high-level DR5 expression, disulfide-mediated clustering, and activation of Caspase 8-Caspase 3 mediated pro-apoptotic signaling. Recognition of the extracellular domain of DR5 by various antibodies is strongly influenced by the pattern of DR5 disulfide bonding, which has important implications for the use of agonistic DR5 antibodies for cancer therapy. Disulfide-defective DR5 mutants do not activate the ER stress response or stimulate autophagy, indicating that these DDA-mediated responses are separable from DR5 activation and pro-apoptotic signaling. Importantly, other ER stressors, including Thapsigargin and Tunicamycin also alter DR5 disulfide bonding in various cancer cell lines and in some instances, DR5 mis-disulfide bonding is potentiated by overriding the Integrated Stress Response (ISR) with inhibitors of the PERK kinase or the ISR inhibitor ISRIB. These observations indicate that the pattern of DR5 disulfide bonding functions as a sensor of ER stress and serves as an effector of proteotoxic stress by driving extrinsic apoptosis independently of extracellular ligands.
Collapse
|
7
|
Wu M, Maiorano G, Stadnicka K. Protein profiles in the transfected oviductal secreting cells of laying hen (Gallus gallus domesticus). Poult Sci 2024; 103:103305. [PMID: 38198917 PMCID: PMC10792652 DOI: 10.1016/j.psj.2023.103305] [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: 08/18/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 01/12/2024] Open
Abstract
Due to the intensive development of novel biopharming applications, there is a need for the in vitro verification models prior to in vivo testing. Laying hen has been already applied as an animal bioreactor to produce the therapeutical enzyme in a rare disease called lysosomal acid lipase deficiency. In this study, we aimed to verify how the proteome of the transfected oviduct epithelial cells would be affected by genetic nonviral modification with the human exogene. The study was based on a previously developed method to cultivate chicken oviduct epithelial cells (COEC). The typical characteristics of the COEC epithelial cells were retained across the experiments. The mean efficiency of nucleofection ranged from 2.6 to 19.7% depending on the cells' isolation and location in the oviduct (upper, infundibulum site, or magnum). The PCR confirmed the incorporation of human interferon alpha2a (hIFNα2a) exogene into the nucleofected COEC but, the production of hIFNα2a protein did not exceed the detection level in this study. The ovalbumin protein was detected in the nontransfected and transfected COEC, which confirmed the normal secreting functions of the cells subject to modification. Proteomic analysis revealed an increase in abundance of the cell adhesion molecules and collagen molecules after introducing gene under ovalbumin promoter. According to the bioinformatic analyses there was a limited negative impact of transfection on cells, and the normal biochemical pathways were not severely disordered. In conclusion, the observations provide new knowledge about the proteomic profile of the manipulated COEC with regard to the retained normal functionality of the cells, which can be informative for avian biopharma research.
Collapse
Affiliation(s)
- Mengjun Wu
- Department of Agricultural, Environmental and Food Sciences, University of Molise, 86100 Campobasso, Italy
| | - Giuseppe Maiorano
- Department of Agricultural, Environmental and Food Sciences, University of Molise, 86100 Campobasso, Italy
| | - Katarzyna Stadnicka
- Faculty of Health Sciences, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-821 Bydgoszcz, Poland.
| |
Collapse
|
8
|
Kim G, Lee J, Ha J, Kang I, Choe W. Endoplasmic Reticulum Stress and Its Impact on Adipogenesis: Molecular Mechanisms Implicated. Nutrients 2023; 15:5082. [PMID: 38140341 PMCID: PMC10745682 DOI: 10.3390/nu15245082] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/30/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Endoplasmic reticulum (ER) stress plays a pivotal role in adipogenesis, which encompasses the differentiation of adipocytes and lipid accumulation. Sustained ER stress has the potential to disrupt the signaling of the unfolded protein response (UPR), thereby influencing adipogenesis. This comprehensive review illuminates the molecular mechanisms that underpin the interplay between ER stress and adipogenesis. We delve into the dysregulation of UPR pathways, namely, IRE1-XBP1, PERK and ATF6 in relation to adipocyte differentiation, lipid metabolism, and tissue inflammation. Moreover, we scrutinize how ER stress impacts key adipogenic transcription factors such as proliferator-activated receptor γ (PPARγ) and CCAAT-enhancer-binding proteins (C/EBPs) along with their interaction with other signaling pathways. The cellular ramifications include alterations in lipid metabolism, dysregulation of adipokines, and aged adipose tissue inflammation. We also discuss the potential roles the molecular chaperones cyclophilin A and cyclophilin B play in adipogenesis. By shedding light on the intricate relationship between ER stress and adipogenesis, this review paves the way for devising innovative therapeutic interventions.
Collapse
Affiliation(s)
- Gyuhui Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jiyoon Lee
- Department of Biological Sciences, Franklin College of Arts and Sciences, University of Georgia, Athens, GA 30609, USA;
| | - Joohun Ha
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
9
|
Cheng KP, Shen WX, Jiang YY, Chen Y, Chen YZ, Tan Y. Deep learning of 2D-Restructured gene expression representations for improved low-sample therapeutic response prediction. Comput Biol Med 2023; 164:107245. [PMID: 37480677 DOI: 10.1016/j.compbiomed.2023.107245] [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: 04/26/2023] [Revised: 06/27/2023] [Accepted: 07/07/2023] [Indexed: 07/24/2023]
Abstract
Clinical outcome prediction is important for stratified therapeutics. Machine learning (ML) and deep learning (DL) methods facilitate therapeutic response prediction from transcriptomic profiles of cells and clinical samples. Clinical transcriptomic DL is challenged by the low-sample sizes (34-286 subjects), high-dimensionality (up to 21,653 genes) and unordered nature of clinical transcriptomic data. The established methods rely on ML algorithms at accuracy levels of 0.6-0.8 AUC/ACC values. Low-sample DL algorithms are needed for enhanced prediction capability. Here, an unsupervised manifold-guided algorithm was employed for restructuring transcriptomic data into ordered image-like 2D-representations, followed by efficient DL of these 2D-representations with deep ConvNets. Our DL models significantly outperformed the state-of-the-art (SOTA) ML models on 82% of 17 low-sample benchmark datasets (53% with >0.05 AUC/ACC improvement). They are more robust than the SOTA models in cross-cohort prediction tasks, and in identifying robust biomarkers and response-dependent variational patterns consistent with experimental indications.
Collapse
Affiliation(s)
- Kai Ping Cheng
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, PR China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, PR China
| | - Wan Xiang Shen
- Bioinformatics and Drug Design Group, Department of Pharmacy, Center for Computational Science and Engineering, National University of Singapore, 117543, Singapore
| | - Yu Yang Jiang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, PR China
| | - Yan Chen
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, PR China
| | - Yu Zong Chen
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, PR China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, PR China.
| | - Ying Tan
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, PR China; The Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, PR China; Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen, 518110, PR China.
| |
Collapse
|
10
|
Gegunde S, Alfonso A, Cifuentes JM, Alvariño R, Pérez-Fuentes N, Vieytes MR, Botana LM. Cyclophilins modify their profile depending on the organ or tissue in a murine inflammatory model. Int Immunopharmacol 2023; 120:110351. [PMID: 37235965 DOI: 10.1016/j.intimp.2023.110351] [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: 02/15/2023] [Revised: 05/10/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023]
Abstract
Inflammation is the leading subjacent cause of many chronic diseases. Despite several studies in the last decades, the molecular mechanism involving its pathophysiology is not fully known. Recently, the implication of cyclophilins in inflammatory-based diseases has been demonstrated. However, the main role of cyclophilins in these processes remains elusive. Hence, a mouse model of systemic inflammation was used to better understand the relationship between cyclophilins and their tissue distribution. To induce inflammation, mice were fed with high-fat diet for 10 weeks. In these conditions, serum levels of interleukins 2 and 6, tumour necrosis factor-α, interferon-ϒ, and the monocyte chemoattractant protein 1 were elevated, evidencing a systemic inflammatory state. Then, in this inflammatory model, cyclophilins and CD147 profiles in the aorta, liver, and kidney were studied. The results demonstrate that, upon inflammatory conditions, cyclophilins A and C expression levels were increased in the aorta. Cyclophilins A and D were augmented in the liver, meanwhile, cyclophilins B and C were diminished. In the kidney, cyclophilins B and C levels were elevated. Furthermore, CD147 receptor was also increased in the aorta, liver, and kidney. In addition, when cyclophilin A was modulated, serum levels of inflammatory mediators were decreased, indicating a reduction in systemic inflammation. Besides, the expression levels of cyclophilin A and CD147 were also reduced in the aorta and liver, when cyclophilin A was modulated. Therefore, these results suggest that each cyclophilin has a different profile depending on the tissue, under inflammatory conditions.
Collapse
Affiliation(s)
- Sandra Gegunde
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain; Grupo de investigación Biodiscovery (IDIS), Lugo, Spain
| | - Amparo Alfonso
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain; Grupo de investigación Biodiscovery (IDIS), Lugo, Spain.
| | - J Manuel Cifuentes
- Departamento de Anatomía, Producción Animal y Ciencias Clínicas Veterinarias, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Rebeca Alvariño
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain; Grupo de investigación Biodiscovery (IDIS), Lugo, Spain
| | - Nadia Pérez-Fuentes
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain; Grupo de investigación Biodiscovery (IDIS), Lugo, Spain
| | - Mercedes R Vieytes
- Departamento de Fisiología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Luis M Botana
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain; Grupo de investigación Biodiscovery (IDIS), Lugo, Spain.
| |
Collapse
|
11
|
Kucharski M, Wirjanata G, Nayak S, Boentoro J, Dziekan JM, Assisi C, van der Pluijm RW, Miotto O, Mok S, Dondorp AM, Bozdech Z. Short tandem repeat polymorphism in the promoter region of cyclophilin 19B drives its transcriptional upregulation and contributes to drug resistance in the malaria parasite Plasmodium falciparum. PLoS Pathog 2023; 19:e1011118. [PMID: 36696458 PMCID: PMC9901795 DOI: 10.1371/journal.ppat.1011118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 02/06/2023] [Accepted: 01/11/2023] [Indexed: 01/26/2023] Open
Abstract
Resistance of the human malaria parasites, Plasmodium falciparum, to artemisinins is now fully established in Southeast Asia and is gradually emerging in Sub-Saharan Africa. Although nonsynonymous SNPs in the pfk13 Kelch-repeat propeller (KREP) domain are clearly associated with artemisinin resistance, their functional relevance requires cooperation with other genetic factors/alterations of the P. falciparum genome, collectively referred to as genetic background. Here we provide experimental evidence that P. falciparum cyclophilin 19B (PfCYP19B) may represent one putative factor in this genetic background, contributing to artemisinin resistance via its increased expression. We show that overexpression of PfCYP19B in vitro drives limited but significant resistance to not only artemisinin but also piperaquine, an important partner drug in artemisinin-based combination therapies. We showed that PfCYP19B acts as a negative regulator of the integrated stress response (ISR) pathway by modulating levels of phosphorylated eIF2α (eIF2α-P). Curiously, artemisinin and piperaquine affect eIF2α-P in an inverse direction that in both cases can be modulated by PfCYP19B towards resistance. Here we also provide evidence that the upregulation of PfCYP19B in the drug-resistant parasites appears to be maintained by a short tandem repeat (SRT) sequence polymorphism in the gene's promoter region. These results support a model that artemisinin (and other drugs) resistance mechanisms are complex genetic traits being contributed to by altered expression of multiple genes driven by genetic polymorphism at their promoter regions.
Collapse
Affiliation(s)
- Michal Kucharski
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Grennady Wirjanata
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Sourav Nayak
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Josephine Boentoro
- School of Biological Sciences, Nanyang Technological University, Singapore
| | | | - Christina Assisi
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Rob W. van der Pluijm
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Olivo Miotto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Sachel Mok
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Arjen M. Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
12
|
Clark V, Waters K, Orsburn B, Bumpus NN, Kundu N, Sczepanski JT, Ray P, Arroyo‐Currás N. Human Cyclophilin B Nuclease Activity Revealed via Nucleic Acid-Based Electrochemical Sensors. Angew Chem Int Ed Engl 2022; 61:e202211292. [PMID: 35999181 PMCID: PMC9633453 DOI: 10.1002/anie.202211292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Indexed: 01/12/2023]
Abstract
Human cyclophilin B (CypB) is oversecreted by pancreatic cancer cells, making it a potential biomarker for early-stage disease diagnosis. Our group is motivated to develop aptamer-based assays to measure CypB levels in biofluids. However, human cyclophilins have been postulated to have collateral nuclease activity, which could impede the use of aptamers for CypB detection. To establish if CypB can hydrolyze electrode-bound nucleic acids, we used ultrasensitive electrochemical sensors to measure CypB's hydrolytic activity. Our sensors use ssDNA and dsDNA in the biologically predominant d-DNA form, and in the nuclease resistant l-DNA form. Challenging such sensors with CypB and control proteins, we unequivocally demonstrate that CypB can cleave nucleic acids. To our knowledge, this is the first study to use electrochemical biosensors to reveal the hydrolytic activity of a protein that is not known to be a nuclease. Future development of CypB bioassays will require the use of nuclease-resistant aptamer sequences.
Collapse
Affiliation(s)
- Vincent Clark
- Chemistry-Biology Interface ProgramZanvyl Krieger School of Arts & SciencesJohns Hopkins UniversityBaltimoreMD 21218USA
| | - Kelly Waters
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMD 21205USA
| | - Ben Orsburn
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMD 21205USA
| | - Namandjé N. Bumpus
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMD 21205USA
| | - Nandini Kundu
- Department of ChemistryTexas A&M University College StationTexasTX 77842USA
| | | | - Partha Ray
- Department of SurgeryDivision of Surgical OncologyMoores Cancer CenterDepartment of MedicineDivision of Infectious Diseases and Global Public HealthUniversity of California San Diego HealthSan DiegoCA 92093USA
| | - Netzahualcóyotl Arroyo‐Currás
- Chemistry-Biology Interface ProgramZanvyl Krieger School of Arts & SciencesJohns Hopkins UniversityBaltimoreMD 21218USA
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMD 21205USA
| |
Collapse
|
13
|
Hill SE, Esquivel AR, Ospina SR, Rahal LM, Dickey CA, Blair LJ. Chaperoning activity of the cyclophilin family prevents tau aggregation. Protein Sci 2022; 31:e4448. [PMID: 36305768 PMCID: PMC9597375 DOI: 10.1002/pro.4448] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/18/2022] [Accepted: 09/11/2022] [Indexed: 11/09/2022]
Abstract
Tauopathies, such as Alzheimer's disease, are characterized by the misfolding and progressive accumulation of the microtubule associated protein tau. Chaperones, tasked with maintaining protein homeostasis, can become imbalanced with age and contribute to the progression of neurodegenerative disease. Cyclophilins are a promising pool of underinvestigated chaperones with peptidyl-prolyl isomerase activity that may play protective roles in regulating tau aggregation. Using a Thioflavin T fluorescence-based assay to monitor in vitro tau aggregation, all eight cyclophilins, which include PPIA to PPIH prevent tau aggregation, with PPIB, PPIC, PPID, and PPIH showing the greatest inhibition. The low thermal stability of PPID and the strong heparin binding of PPIB undermines the simplistic interpretation of reduced tau aggregation. In a cellular model of tau accumulation, all cyclophilins, except PPID and PPIH, reduce insoluble tau. PPIB, PPIC, PPIE, and PPIF also reduce soluble tau levels with PPIC exclusively protecting cells from tau seeding. Overall, this study demonstrates cyclophilins prevent tau fibril formation and many reduce cellular insoluble tau accumulation with PPIC having the greatest potential as a molecular tool to mitigate tau seeding and accumulation.
Collapse
Affiliation(s)
- Shannon E. Hill
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Abigail R. Esquivel
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Santiago Rodriguez Ospina
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Lauren M. Rahal
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Chad A. Dickey
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Laura J. Blair
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
- Research ServiceJames A. Haley Veterans HospitalTampaFloridaUSA
| |
Collapse
|
14
|
The role of cyclophilins in viral infec and the immune response. J Infect 2022; 85:365-373. [PMID: 35934139 DOI: 10.1016/j.jinf.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022]
|
15
|
Alvariño R, Alfonso A, Pech-Puch D, Gegunde S, Rodríguez J, Vieytes MR, Jiménez C, Botana LM. Furanoditerpenes from Spongia (Spongia) tubulifera Display Mitochondrial-Mediated Neuroprotective Effects by Targeting Cyclophilin D. ACS Chem Neurosci 2022; 13:2449-2463. [PMID: 35901231 PMCID: PMC9686139 DOI: 10.1021/acschemneuro.2c00208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Neuroprotective properties of five previously described furanoditerpenes 1-5, isolated from Spongia (Spongia) tubulifera, were evaluated in an in vitro oxidative stress model in SH-SY5Y cells. Dose-response treatments revealed that 1-5 improved cell survival at nanomolar concentrations through the restoration of mitochondrial membrane potential and the reduction of reactive oxygen species. Their ability to prevent the mitochondrial permeability transition pore opening was also assessed, finding that 4 and 5 inhibited the channel at 0.001 μM. This inhibition was accompanied by a decrease in the expression of cyclophilin D, the main regulator of the pore, which was also reduced by 1 and 2. However, the activation of ERK and GSK3β, upstream modulators of the channel, was not affected by compounds. Therefore, their ability to bind cyclophilin D was evaluated by surface plasmon resonance, observing that 2-5 presented equilibrium dissociation constants in the micromolar range. All compounds also showed affinity for cyclophilin A, being 1 selective toward this isoform, while 2 and 5 exhibited selectivity for cyclophilin D. When the effects on the intracellular expression of cyclophilins A-C were determined, it was found that only 1 decreased cyclophilin A, while cyclophilins B and C were diminished by most compounds, displaying enhanced effects under oxidative stress conditions. Results indicate that furanoditerpenes 1-5 have mitochondrial-mediated neuroprotective properties through direct interaction with cyclophilin D. Due to the important role of this protein in oxidative stress and inflammation, compounds are promising drugs for new therapeutic strategies against neurodegeneration.
Collapse
Affiliation(s)
- Rebeca Alvariño
- Departamento
de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain,Grupo
Investigación Biodiscovery, IDIS, 27002 Lugo, Spain
| | - Amparo Alfonso
- Departamento
de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain,Grupo
Investigación Biodiscovery, IDIS, 27002 Lugo, Spain
| | - Dawrin Pech-Puch
- Centro
de Investigacións Científicas Avanzadas (CICA) e Departamento
de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Spain,Departamento
de Biología Marina, Campus de Ciencias Biológicas y
Agropecuarias, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Yucatán, 97100 Mérida, Yucatán, Mexico
| | - Sandra Gegunde
- Departamento
de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain,Grupo
Investigación Biodiscovery, IDIS, 27002 Lugo, Spain,Fundación
Instituto de Investigación Sanitario Santiago de Compostela
(FIDIS), Hospital Universitario Lucus Augusti, 27002 Lugo, Spain
| | - Jaime Rodríguez
- Centro
de Investigacións Científicas Avanzadas (CICA) e Departamento
de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Spain
| | - Mercedes R. Vieytes
- Grupo
Investigación Biodiscovery, IDIS, 27002 Lugo, Spain,Departamento
de Fisiología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Carlos Jiménez
- Centro
de Investigacións Científicas Avanzadas (CICA) e Departamento
de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Spain,. Phone/Fax: +34881012170
| | - Luis M. Botana
- Departamento
de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain,Grupo
Investigación Biodiscovery, IDIS, 27002 Lugo, Spain,. Phone/Fax: +34982822233
| |
Collapse
|
16
|
Yang X, Shu B, Zhou Y, Li Z, He C. Ppic modulates CCl 4-induced liver fibrosis and TGF-β-caused mouse hepatic stellate cell activation and regulated by miR-137-3p. Toxicol Lett 2021; 350:52-61. [PMID: 34224798 DOI: 10.1016/j.toxlet.2021.06.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/10/2021] [Accepted: 06/30/2021] [Indexed: 11/28/2022]
Abstract
Hepatic stellate cell activation, characterized by hyperproliferation and increased release of collagens, is a critical event during the initiation and development of hepatic fibrosis. The deregulated genes among different expression profiles based on online datasets were analyzed, attempting to identify novel potential biomarkers and treatment targets for hepatic fibrosis. The abnormal upregulation of mouse peptidylprolyl isomerase C (Ppic) within the CCl4-caused hepatic fibrosis model in mice was identified according to bioinformatics and experimental analyses. The knockdown of Ppic in the CCl4-caused liver fibrosis murine model significantly improved CCl4-caused liver damage, decreased the fibrotic area, reduced ECM deposition, and reduced the hydroxyproline levels. The knockdown of Ppic in TGF-β-stimulated mouse hepatic stellate cells inhibited cell proliferation and decreased ECM levels. Through direct targeting, miR-137-3p negatively regulated Ppic expression. Contrastingly to Ppic knockdown, miR-137-3p inhibition further promoted cell proliferation and boosted ECM levels; the effects of miR-137-3p inhibition could be partially reversed by Ppic knockdown. Altogether, mmu-miR-137-3p directly targets Ppic and forms a regulatory axis with Ppic, modulating CCl4-caused hepatic fibrosis in mice and TGF-β-caused mouse hepatic stellate cell activation.
Collapse
Affiliation(s)
- Xin Yang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Bo Shu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yingxia Zhou
- Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Zhuan Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, 410013, China; Department of Pharmacy, Hunan Normal University School of Medicine, Changsha, Hunan, 410013, China
| | - Chao He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
17
|
Paiva RS, Ramos CV, Azenha SR, Alves C, Basto AP, Graca L, Martins VC. Peptidylprolyl isomerase C (Ppic) regulates invariant Natural Killer T cell (iNKT) differentiation in mice. Eur J Immunol 2021; 51:1968-1979. [PMID: 33864384 PMCID: PMC8453708 DOI: 10.1002/eji.202048924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 03/18/2021] [Accepted: 04/13/2021] [Indexed: 12/25/2022]
Abstract
Peptidyl‐prolyl cis‐trans isomerase C (Ppic) is expressed in several bone marrow (BM) hematopoietic progenitors and in T‐cell precursors. Since the expression profile of Ppic in the hematoimmune system was suggestive that it could play a role in hematopoiesis and/or T lymphocyte differentiation, we sought to test that hypothesis in vivo. Specifically, we generated a Ppic‐deficient mouse model by targeting the endogenous locus by CRISPR/Cas9 and tested the requirement of Ppic in hematopoiesis. Several immune cell lineages covering BM progenitors, lymphocyte precursors, as well as mature cells at the periphery were analyzed. While most lineages were unaffected, invariant NKT (iNKT) cells were reduced in percentage and absolute cell numbers in the Ppic‐deficient thymus. This affected the most mature stages in the thymus, S2 and S3, and the phenotype was maintained at the periphery. Additionally, immature transitional T1 and T2 B lymphocytes were increased in the Ppic‐deficient spleen, but the phenotype was lost in mature B lymphocytes. In sum, our data show that Ppic is dispensable for myeloid cells, platelets, erythrocytes, αβ, and γδ T lymphocytes in vivo in the steady state, while being involved in B‐ and iNKT cell differentiation.
Collapse
Affiliation(s)
- Ricardo S Paiva
- Lymphocyte Development and Leukemogenesis Laboratory, Instituto Gulbenkian de Ciência, Calouste Gulbenkian Foundation, Oeiras, Portugal
| | - Camila V Ramos
- Lymphocyte Development and Leukemogenesis Laboratory, Instituto Gulbenkian de Ciência, Calouste Gulbenkian Foundation, Oeiras, Portugal
| | - Sara R Azenha
- Lymphocyte Development and Leukemogenesis Laboratory, Instituto Gulbenkian de Ciência, Calouste Gulbenkian Foundation, Oeiras, Portugal
| | - Carolina Alves
- Lymphocyte Development and Leukemogenesis Laboratory, Instituto Gulbenkian de Ciência, Calouste Gulbenkian Foundation, Oeiras, Portugal
| | - Afonso P Basto
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisboa, Portugal
| | - Luis Graca
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Vera C Martins
- Lymphocyte Development and Leukemogenesis Laboratory, Instituto Gulbenkian de Ciência, Calouste Gulbenkian Foundation, Oeiras, Portugal
| |
Collapse
|
18
|
Gegunde S, Alfonso A, Alvariño R, Alonso E, Botana LM. Cyclophilins A, B, and C Role in Human T Lymphocytes Upon Inflammatory Conditions. Front Immunol 2021; 12:609196. [PMID: 33859635 PMCID: PMC8042163 DOI: 10.3389/fimmu.2021.609196] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/15/2021] [Indexed: 02/01/2023] Open
Abstract
Cyclophilins (Cyps) are a group of peptidyl-prolyl cis/trans isomerases that play crucial roles in regulatory mechanisms of cellular physiology and pathology in several inflammatory conditions. Their receptor, CD147, also participates in the development and progression of the inflammatory response. Nevertheless, the main function of Cyps and their receptor are yet to be deciphered. The release of CypA and the expression of the CD147 receptor in activated T lymphocytes were already described, however, no data are available about other Cyps in these cells. Therefore, in the present work intra and extracellular CypA, B and C levels were measured followed by induced inflammatory conditions. After activation of T lymphocytes by incubation with concanavalin A, both intra and extracellular Cyps levels and the CD147 membrane receptor expression were increased leading to cell migration towards circulating CypA and CypB as chemoattractants. When CypA was modulated by natural and synthetic compounds, the inflammatory cascade was avoided including T cell migration. Our results strengthen the relationship between CypA, B, and C, their receptor, and the inflammatory process in human T lymphocytes, associating CypC with these cells for the first time.
Collapse
Affiliation(s)
- Sandra Gegunde
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, Spain.,Grupo Investigación Biodiscovery, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Amparo Alfonso
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, Spain.,Grupo Investigación Biodiscovery, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Rebeca Alvariño
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, Spain.,Grupo Investigación Biodiscovery, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Eva Alonso
- Grupo Investigación Biodiscovery, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Luis M Botana
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, Spain.,Grupo Investigación Biodiscovery, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
19
|
Cross-Platform Comparison of Computer-assisted Image Analysis Quantification of In Situ mRNA Hybridization in Investigative Pathology. Appl Immunohistochem Mol Morphol 2020; 27:15-26. [PMID: 28682833 DOI: 10.1097/pai.0000000000000542] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although availability of automated platforms has proliferated, there is no standard practice for computer-assisted generation of scores for mRNA in situ hybridization (ISH) visualized by brightfield microscopic imaging on tissue sections. To address this systematically, an ISH for peptidylprolyl isomerase B (PPIB) (cyclophilin B) mRNA was optimized and applied to a tissue microarray of archival non-small cell lung carcinoma cases, and then automated image analysis for PPIB was refined across 4 commercially available software platforms. Operator experience and scoring results from ImageScope, HALO, CellMap, and Developer XD were systematically compared with each other and to manual pathologist scoring. Markup images were compared and contrasted for accuracy, the ability of the platform to identify cells, and the ease of visual assessment to determine appropriate interpretation. Comparing weighted scoring approaches using H-scores (Developer XD, ImageScope, and manual scoring) a correlation was observed (R value=0.7955), and association between the remaining 2 approaches (HALO and CellMap) was of similar value. ImageScope showed the highest R value in comparison with manual scoring (0.7377). Mean-difference plots showed that HALO produced the highest relative normalized values, suggesting higher relative sensitivity. ImageScope overestimated PPIB ISH signal at the high end of the range scores; however, this tendency was not observed in other platforms. HALO emerged with the highest number of favorable observations, no apparent systematic bias in score generation compared with the other methods, and potentially higher sensitivity to detect ISH. HALO may serve as a tool to empower teams of investigative pathology laboratory scientists to assist pathologists readily with quantitative scoring of ISH.
Collapse
|
20
|
Lizák B, Birk J, Zana M, Kosztyi G, Kratschmar DV, Odermatt A, Zimmermann R, Geiszt M, Appenzeller-Herzog C, Bánhegyi G. Ca 2+ mobilization-dependent reduction of the endoplasmic reticulum lumen is due to influx of cytosolic glutathione. BMC Biol 2020; 18:19. [PMID: 32101139 PMCID: PMC7043043 DOI: 10.1186/s12915-020-0749-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/11/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The lumen of the endoplasmic reticulum (ER) acts as a cellular Ca2+ store and a site for oxidative protein folding, which is controlled by the reduced glutathione (GSH) and glutathione-disulfide (GSSG) redox pair. Although depletion of luminal Ca2+ from the ER provokes a rapid and reversible shift towards a more reducing poise in the ER, the underlying molecular basis remains unclear. RESULTS We found that Ca2+ mobilization-dependent ER luminal reduction was sensitive to inhibition of GSH synthesis or dilution of cytosolic GSH by selective permeabilization of the plasma membrane. A glutathione-centered mechanism was further indicated by increased ER luminal glutathione levels in response to Ca2+ efflux. Inducible reduction of the ER lumen by GSH flux was independent of the Ca2+-binding chaperone calreticulin, which has previously been implicated in this process. However, opening the translocon channel by puromycin or addition of cyclosporine A mimicked the GSH-related effect of Ca2+ mobilization. While the action of puromycin was ascribable to Ca2+ leakage from the ER, the mechanism of cyclosporine A-induced GSH flux was independent of calcineurin and cyclophilins A and B and remained unclear. CONCLUSIONS Our data strongly suggest that ER influx of cytosolic GSH, rather than inhibition of local oxidoreductases, is responsible for the reductive shift upon Ca2+ mobilization. We postulate the existence of a Ca2+- and cyclosporine A-sensitive GSH transporter in the ER membrane. These findings have important implications for ER redox homeostasis under normal physiology and ER stress.
Collapse
Affiliation(s)
- Beáta Lizák
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary.
| | - Julia Birk
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Melinda Zana
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- "Momentum" Peroxidase Enzyme Research Group of the Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary
| | - Gergely Kosztyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Denise V Kratschmar
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Richard Zimmermann
- Medical Biochemistry and Molecular Biology, Saarland University, 66421, Homburg, Germany
| | - Miklós Geiszt
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- "Momentum" Peroxidase Enzyme Research Group of the Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary
| | - Christian Appenzeller-Herzog
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
- University Medical Library, University of Basel, Spiegelgasse 5, 4051, Basel, Switzerland.
| | - Gábor Bánhegyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
21
|
Alfonso A, Bayón J, Gegunde S, Alonso E, Alvariño R, Santás-Álvarez M, Testa-Fernández A, Rios-Vázquez R, González-Juanatey C, Botana LM. High Serum Cyclophilin C levels as a risk factor marker for Coronary Artery Disease. Sci Rep 2019; 9:10576. [PMID: 31332225 PMCID: PMC6646393 DOI: 10.1038/s41598-019-46988-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 07/09/2019] [Indexed: 12/12/2022] Open
Abstract
Cyclophilins (Cyps) are ubiquitous proteins that belong to the immunophilins family consistently associated with inflammatory and cardiovascular diseases. While levels of CypA have been extensively studied, less data are available for other Cyps. The purpose of this case-control study was to determine the relationship of Cyps (A, B, C and D) with coronary artery disease (CAD) and eight inflammation markers. Serum levels of Cyps, interleukins and metalloproteinases were measured in serum collected from 84 subjects. Participants were divided into two sub-groups based on CAD diagnosis: 40 CAD patients and 44 control volunteers. Serum levels of CypA, CypB and CypC, IL-1β and IL-6 were significantly higher in CAD patients. Bivariate correlation analysis revealed a significant positive correlation between Cyps and several blood and biochemical parameters. When the ability of Cyps levels for CAD diagnosis was evaluated, higher sensitivity and selectivity values were obtained with CypC (c-statistic 0.891, p < 0.001) indicating that it is a good marker of CAD disease, while less conclusive results were obtained with CypA (c-statistic 0.748, p < 0.001) and CypB (c-statistic 0.655, p < 0.014). In addition, significant correlations of traditional CAD risk factors and CypC were observed. In summary, high levels of CypC are a risk factor for CAD and therefore it can be proposed as a new biomarker for this disease.
Collapse
Affiliation(s)
- Amparo Alfonso
- Pharmacology Department, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002, Lugo, Spain.
| | - Jeremías Bayón
- Cardiology Department, Hospital Universitario Lucus Augusti, 27003, Lugo, Spain
| | - Sandra Gegunde
- Pharmacology Department, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002, Lugo, Spain
| | - Eva Alonso
- Pharmacology Department, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002, Lugo, Spain
| | - Rebeca Alvariño
- Pharmacology Department, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002, Lugo, Spain
| | | | - Ana Testa-Fernández
- Cardiology Department, Hospital Universitario Lucus Augusti, 27003, Lugo, Spain
| | - Ramón Rios-Vázquez
- Cardiology Department, Hospital Universitario Lucus Augusti, 27003, Lugo, Spain
| | | | - Luis M Botana
- Pharmacology Department, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002, Lugo, Spain
| |
Collapse
|
22
|
Gómez-Fernández P, Urtasun A, Astobiza I, Mena J, Alloza I, Vandenbroeck K. Pharmacological Targeting of the ER-Resident Chaperones GRP94 or Cyclophilin B Induces Secretion of IL-22 Binding Protein Isoform-1 (IL-22BPi1). Int J Mol Sci 2019; 20:ijms20102440. [PMID: 31108847 PMCID: PMC6566634 DOI: 10.3390/ijms20102440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 01/05/2023] Open
Abstract
Of the three interleukin-22 binding protein (IL-22BP) isoforms produced by the human IL22RA2 gene, IL-22BPi2 and IL-22BPi3 are capable of neutralizing IL-22. The longest isoform, IL-22BPi1, does not bind IL-22, is poorly secreted, and its retention within the endoplasmic reticulum (ER) is associated with induction of an unfolded protein response (UPR). Therapeutic modulation of IL-22BPi2 and IL-22BPi3 production may be beneficial in IL-22-dependent disorders. Recently, we identified the ER chaperones GRP94 and cyclophilin B in the interactomes of both IL-22BPi1 and IL-22BPi2. In this study, we investigated whether secretion of the IL-22BP isoforms could be modulated by pharmacological targeting of GRP94 and cyclophilin B, either by means of geldanamycin, that binds to the ADP/ATP pocket shared by HSP90 paralogs, or by cyclosporin A, which causes depletion of ER cyclophilin B levels through secretion. We found that geldanamycin and its analogs did not influence secretion of IL-22BPi2 or IL-22BPi3, but significantly enhanced intracellular and secreted levels of IL-22BPi1. The secreted protein was heterogeneously glycosylated, with both high-mannose and complex-type glycoforms present. In addition, cyclosporine A augmented the secretion of IL-22BPi1 and reduced that of IL-22BPi2 and IL-22BPi3. Our data indicate that the ATPase activity of GRP94 and cyclophilin B are instrumental in ER sequestration and degradation of IL-22BPi1, and that blocking these factors mobilizes IL-22BPi1 toward the secretory route.
Collapse
Affiliation(s)
- Paloma Gómez-Fernández
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
| | - Andoni Urtasun
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
| | - Ianire Astobiza
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
| | - Jorge Mena
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
| | - Iraide Alloza
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
| | - Koen Vandenbroeck
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48490 Leioa, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
23
|
Wang M, Ferreira RB, Law ME, Davis BJ, Yaaghubi E, Ghilardi AF, Sharma A, Avery BA, Rodriguez E, Chiang CW, Narayan S, Heldermon CD, Castellano RK, Law BK. A novel proteotoxic combination therapy for EGFR+ and HER2+ cancers. Oncogene 2019; 38:4264-4282. [PMID: 30718919 DOI: 10.1038/s41388-019-0717-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 02/08/2023]
Abstract
While HER2 and EGFR are overexpressed in breast cancers and multiple other types of tumors, the use of EGFR and/or HER2 inhibitors have failed to cure many cancer patients, largely because cancers acquire resistance to HER2/EGFR-specific drugs. Cancers that overexpress the HER-family proteins EGFR, HER2, and HER3 are uniquely sensitive to agents that disrupt HER2 and EGFR protein folding. We previously showed that disruption of disulfide bond formation by Disulfide Disrupting Agents (DDAs) kills HER2/EGFR overexpressing cells through multiple mechanisms. Herein, we show that interference with proline isomerization in HER2/EGFR overexpressing cells also induces cancer cell death. The peptidyl-prolyl isomerase inhibitor Cyclosporine A (CsA) selectively kills EGFR+ or HER2+ breast cancer cells in vitro by activating caspase-dependent apoptotic pathways. Further, CsA synergizes with the DDA tcyDTDO to kill HER2/EGFR overexpressing cells in vitro and the two agents cooperate to kill HER2+ tumors in vivo. There is a critical need for novel strategies to target HER2+ and EGFR+ cancers that are resistant to currently available mechanism-based agents. Drugs that target HER2/EGFR protein folding, including DDAs and CsA, have the potential to kill cancers that overexpress EGFR or HER2 through the induction of proteostatic synthetic lethality.
Collapse
Affiliation(s)
- Mengxiong Wang
- Department of Pharmacology, University of Florida, Gainesville, FL, 32610, USA
| | - Renan B Ferreira
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
| | - Mary E Law
- Department of Pharmacology, University of Florida, Gainesville, FL, 32610, USA
| | - Bradley J Davis
- Department of Pharmacology, University of Florida, Gainesville, FL, 32610, USA
| | - Elham Yaaghubi
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
| | - Amanda F Ghilardi
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
| | - Abhisheak Sharma
- Department of Pharmaceutics, University of Florida, Gainesville, FL, 32610, USA
| | - Bonnie A Avery
- Department of Pharmaceutics, University of Florida, Gainesville, FL, 32610, USA
| | - Edgardo Rodriguez
- Department of Pharmacology, University of Florida, Gainesville, FL, 32610, USA
| | - Chi-Wu Chiang
- Institute of Molecular Medicine, College of Medicine and Center for Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Satya Narayan
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL, 32610, USA.,UF-Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Coy D Heldermon
- UF-Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA.,Department of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Ronald K Castellano
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA. .,UF-Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA.
| | - Brian K Law
- Department of Pharmacology, University of Florida, Gainesville, FL, 32610, USA. .,UF-Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
24
|
Wang B, Lin L, Wang H, Guo H, Gu Y, Ding W. Overexpressed cyclophilin B suppresses aldosterone-induced proximal tubular cell injury both in vitro and in vivo. Oncotarget 2018; 7:69309-69320. [PMID: 27732567 PMCID: PMC5342479 DOI: 10.18632/oncotarget.12503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/28/2016] [Indexed: 11/25/2022] Open
Abstract
The renin-angiotensin-aldosterone system (RAAS) is overactivated in patients with chronic kidney disease. Oxidative stress and endoplasmic reticulum stress (ERS) are two major mechanisms responsible for aldosterone-induced kidney injury. Cyclophilin (CYP) B is a chaperone protein that accelerates the rate of protein folding through its peptidyl-prolyl cis-trans isomerase (PPIase) activity. We report that overexpression of wild-type CYPB attenuated aldosterone-induced oxidative stress (evidenced by reduced production of reactive oxygen species and improved mitochondrial dysfunction), ERS (indicated by reduced expression of the ERS markers glucose-regulated protein 78 [GRP78] and C/-EBP homologous protein [CHOP]), and tubular cell apoptosis in comparison with aldosterone-induced human kidney-2 (HK-2) cells. The in vivo study also yielded similar results. Hence, CYPB performs a crucial function in protecting cells against aldosterone-induced oxidative stress, ERS, and tubular cell injury via its PPIase activity.
Collapse
Affiliation(s)
- Bin Wang
- Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University, Xuhui, Shanghai, P.R. China
| | - Lilu Lin
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, P.R. China
| | - Haidong Wang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, P.R. China
| | - Honglei Guo
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, P.R. China
| | - Yong Gu
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, P.R. China
| | - Wei Ding
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, P.R. China
| |
Collapse
|
25
|
Abstract
Cysteine thiols are among the most reactive functional groups in proteins, and their pairing in disulfide linkages is a common post-translational modification in proteins entering the secretory pathway. This modest amino acid alteration, the mere removal of a pair of hydrogen atoms from juxtaposed cysteine residues, contrasts with the substantial changes that characterize most other post-translational reactions. However, the wide variety of proteins that contain disulfides, the profound impact of cross-linking on the behavior of the protein polymer, the numerous and diverse players in intracellular pathways for disulfide formation, and the distinct biological settings in which disulfide bond formation can take place belie the simplicity of the process. Here we lay the groundwork for appreciating the mechanisms and consequences of disulfide bond formation in vivo by reviewing chemical principles underlying cysteine pairing and oxidation. We then show how enzymes tune redox-active cofactors and recruit oxidants to improve the specificity and efficiency of disulfide formation. Finally, we discuss disulfide bond formation in a cellular context and identify important principles that contribute to productive thiol oxidation in complex, crowded, dynamic environments.
Collapse
Affiliation(s)
- Deborah Fass
- Department of Structural Biology, Weizmann Institute of Science , Rehovot 7610001, Israel
| | - Colin Thorpe
- Department of Chemistry and Biochemistry, University of Delaware , Newark, Delaware 19716, United States
| |
Collapse
|
26
|
A potential role of the unfolded protein response in post-transplant cancer. Clin Sci (Lond) 2017. [PMID: 28645931 DOI: 10.1042/cs20170152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cancer is one of the major causes of mortality in organ transplant patients receiving immunosuppressive regimen based on Cyclosporin A (CsA). Organ transplantation and chronic immunosuppression are typically associated with skin cancers (both squamous cell carcinoma and melanoma) and renal cell carcinoma (RCC). Recent studies have shown that in addition to its immunosuppressive effects, accounted for by the inhibition of calcineurin and the modulation of the transcriptional programme of lymphocytes, CsA also directly stimulates the growth and aggressive behaviour of various cancer cells. Using renal carcinogenesis as an example, we discuss the current evidence for a role of cellular proteostasis, i.e. the regulation of the production, maturation and turnover of proteins in eukaryotic cells, in tumorigenesis arising under conditions of chronic immunosuppression. We present the recent studies showing that CsA induces the unfolded protein response (UPR) in normal and transformed kidney cells. We examine how the UPR might be important, considering in particular the genomic analyses showing the existence of a correlation between the levels of expression of the actors of the UPR, the chaperones of the endoplasmic reticulum (ER) and the aggressiveness of renal carcinoma. The UPR may offer a possible explanation for how immunosuppressive regimens based on CsA promote renal carcinogenesis. We discuss the opportunities offered by this biological knowledge in terms of screening, diagnosis and treatment of post-transplant cancers, and propose possible future translational studies examining the role of tumour proteostasis and the UPR in this context.
Collapse
|
27
|
Liu ZY, Huang J, Liu NN, Zheng M, Zhao T, Zhao BC, Wang YM, Pu JL. Molecular Mechanisms of Increased Heart Rate in Shenxianshengmai-treated Bradycardia Rabbits. Chin Med J (Engl) 2017; 130:179-186. [PMID: 28091410 PMCID: PMC5282675 DOI: 10.4103/0366-6999.197999] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background: The molecular mechanisms of Shenxianshengmai (SXSM), a traditional Chinese medicine, on bradycardia have been incompletely understood. The study tried to investigate the gene expression profile and proteomics of bradycardia rabbits’ hearts after SXSM treatment. Methods: Twenty-four adult rabbits were randomly assigned in four groups: sham, model, model plus SXSM treatment, and sham plus SXSM treatment groups. Heart rate was recorded in all rabbits. Then, total RNA of atria and proteins of ventricle were isolated and quantified, respectively. Gene expression profiling was conducted by gene expression chip, and quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR) was performed to confirm the results of gene expression chip. We used isobaric tags for elative and absolute quantitation and Western blotting to identify altered proteins after SXSM treatment. Results: There was a constant decrease in the mean heart rate (32%, from 238 ± 6 beats/min to 149 ± 12 beats/min) after six weeks in model compared with that in sham group. This effect was partially reversed by 4-week SXSM treatment. Complementary DNA microarray demonstrated that the increased acetylcholinesterase and reduced nicotinic receptor were take responsibility for the increased heart rate. In addition, proteins involved in calcium handling and signaling were affected by SXSM treatment. Real-time RT-PCR verified the results from gene chip. Results from proteomics demonstrated that SXSM enhanced oxidative phosphorylation and tricarboxylic acid (TCA) cycle in ventricular myocardium to improve ATP generation. Conclusions: Long-term SXSM stimulates sympathetic transmission by increasing the expression of acetylcholinesterase and reduces the expression of nicotinic receptor to increase heart rate. SXSM also restored the calcium handling genes and altered genes involved in signaling. In addition, SXSM improves the ATP supply of ventricular myocardium by increasing proteins involved in TCA cycle and oxidation-respiratory chain.
Collapse
Affiliation(s)
- Zhou-Ying Liu
- State Key Laboratory of Translational Cardiovascular Medicine, Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037; Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Jian Huang
- State Key Laboratory of Translational Cardiovascular Medicine, Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Na-Na Liu
- State Key Laboratory of Translational Cardiovascular Medicine, Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Min Zheng
- State Key Laboratory of Translational Cardiovascular Medicine, Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Tao Zhao
- Shandong Buchang Pharmaceutical Co. Ltd., Heze, Shandong 274003, China
| | - Bu-Chang Zhao
- Shandong Buchang Pharmaceutical Co. Ltd., Heze, Shandong 274003, China
| | - Yi-Min Wang
- Shandong Buchang Pharmaceutical Co. Ltd., Heze, Shandong 274003, China
| | - Jie-Lin Pu
- State Key Laboratory of Translational Cardiovascular Medicine, Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
28
|
Andrews MC, Cursons J, Hurley DG, Anaka M, Cebon JS, Behren A, Crampin EJ. Systems analysis identifies miR-29b regulation of invasiveness in melanoma. Mol Cancer 2016; 15:72. [PMID: 27852308 PMCID: PMC5112703 DOI: 10.1186/s12943-016-0554-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 10/31/2016] [Indexed: 02/08/2023] Open
Abstract
Background In many cancers, microRNAs (miRs) contribute to metastatic progression by modulating phenotypic reprogramming processes such as epithelial-mesenchymal plasticity. This can be driven by miRs targeting multiple mRNA transcripts, inducing regulated changes across large sets of genes. The miR-target databases TargetScan and DIANA-microT predict putative relationships by examining sequence complementarity between miRs and mRNAs. However, it remains a challenge to identify which miR-mRNA interactions are active at endogenous expression levels, and of biological consequence. Methods We developed a workflow to integrate TargetScan and DIANA-microT predictions into the analysis of data-driven associations calculated from transcript abundance (RNASeq) data, specifically the mutual information and Pearson’s correlation metrics. We use this workflow to identify putative relationships of miR-mediated mRNA repression with strong support from both lines of evidence. Applying this approach systematically to a large, published collection of unique melanoma cell lines – the Ludwig Melbourne melanoma (LM-MEL) cell line panel – we identified putative miR-mRNA interactions that may contribute to invasiveness. This guided the selection of interactions of interest for further in vitro validation studies. Results Several miR-mRNA regulatory relationships supported by TargetScan and DIANA-microT demonstrated differential activity across cell lines of varying matrigel invasiveness. Strong negative statistical associations for these putative regulatory relationships were consistent with target mRNA inhibition by the miR, and suggest that differential activity of such miR-mRNA relationships contribute to differences in melanoma invasiveness. Many of these relationships were reflected across the skin cutaneous melanoma TCGA dataset, indicating that these observations also show graded activity across clinical samples. Several of these miRs are implicated in cancer progression (miR-211, -340, -125b, −221, and -29b). The specific role for miR-29b-3p in melanoma has not been well studied. We experimentally validated the predicted miR-29b-3p regulation of LAMC1 and PPIC and LASP1, and show that dysregulation of miR-29b-3p or these mRNA targets can influence cellular invasiveness in vitro. Conclusions This analytic strategy provides a comprehensive, systems-level approach to identify miR-mRNA regulation in high-throughput cancer data, identifies novel putative interactions with functional phenotypic relevance, and can be used to direct experimental resources for subsequent experimental validation. Computational scripts are available: http://github.com/uomsystemsbiology/LMMEL-miR-miner Electronic supplementary material The online version of this article (doi:10.1186/s12943-016-0554-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Miles C Andrews
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, 3084, Australia.,Department of Medicine, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Joseph Cursons
- Systems Biology Laboratory, University of Melbourne, Parkville, VIC, 3010, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science, University of Melbourne, Parkville, VIC, 3010, Australia.,School of Mathematics and Statistics, University of Melbourne, Parkville, VIC, 3010, Australia.,Centre for Systems Genomics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daniel G Hurley
- Systems Biology Laboratory, University of Melbourne, Parkville, VIC, 3010, Australia.,School of Mathematics and Statistics, University of Melbourne, Parkville, VIC, 3010, Australia.,Centre for Systems Genomics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthew Anaka
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, 3084, Australia.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jonathan S Cebon
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia. .,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, 3084, Australia. .,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, 3084, Australia. .,Department of Medicine, University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Andreas Behren
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia. .,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immunobiology Laboratory, Heidelberg, VIC, 3084, Australia. .,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, 3084, Australia.
| | - Edmund J Crampin
- Department of Medicine, University of Melbourne, Parkville, VIC, 3010, Australia. .,Systems Biology Laboratory, University of Melbourne, Parkville, VIC, 3010, Australia. .,ARC Centre of Excellence in Convergent Bio-Nano Science, University of Melbourne, Parkville, VIC, 3010, Australia. .,School of Mathematics and Statistics, University of Melbourne, Parkville, VIC, 3010, Australia. .,Centre for Systems Genomics, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
29
|
Wu FL, Liu WY, Van Poucke S, Braddock M, Jin WM, Xiao J, Li XK, Zheng MH. Targeting endoplasmic reticulum stress in liver disease. Expert Rev Gastroenterol Hepatol 2016; 10:1041-1052. [PMID: 27093595 DOI: 10.1080/17474124.2016.1179575] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The accumulation of unfolded protein in the endoplasmic reticulum (ER) initiates an unfolded protein response (UPR) via three signal transduction cascades, which involve protein kinase RNA-like ER kinase (PERK), inositol requiring enzyme-1α (IRE1α) and activating transcription factor-6α (ATF6α). An ER stress response is observed in nearly all physiologies related to acute and chronic liver disease and therapeutic targeting of the mechanisms implicated in UPR signaling have attracted considerable attention. AREAS COVERED This review focuses on the correlation between ER stress and liver disease and the possible targets which may drive the potential for novel therapeutic intervention. Expert Commentary: We describe pathways which are involved in UPR signaling and their potential correlation with various liver diseases and underlying mechanisms which may present opportunities for novel therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Fa-Ling Wu
- a Department of Hepatology, Liver Research Center , the First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
- b Institute of Hepatology , Wenzhou Medical University , Wenzhou , China
| | - Wen-Yue Liu
- c Department of Endocrinology , the First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| | - Sven Van Poucke
- d Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy , Ziekenhuis Oost-Limburg , Genk , Belgium
| | - Martin Braddock
- e Global Medicines Development , AstraZeneca R&D , Alderley Park , UK
| | - Wei-Min Jin
- f Department of Infection Diseases , People Hospital of Wencheng County , Wenzhou , China
| | - Jian Xiao
- g Institute of Biology Science , Wenzhou University , Wenzhou , China
- h School of Pharmacy , Wenzhou Medical University , Wenzhou , China
| | - Xiao-Kun Li
- g Institute of Biology Science , Wenzhou University , Wenzhou , China
- h School of Pharmacy , Wenzhou Medical University , Wenzhou , China
| | - Ming-Hua Zheng
- a Department of Hepatology, Liver Research Center , the First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
- b Institute of Hepatology , Wenzhou Medical University , Wenzhou , China
| |
Collapse
|
30
|
Dubnikov T, Ben-Gedalya T, Reiner R, Hoepfner D, Cabral WA, Marini JC, Cohen E. PrP-containing aggresomes are cytosolic components of an ER quality control mechanism. J Cell Sci 2016; 129:3635-3647. [PMID: 27550517 DOI: 10.1242/jcs.186981] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 08/13/2016] [Indexed: 11/20/2022] Open
Abstract
Limited detoxification capacity often directs aggregation-prone, potentially hazardous, misfolded proteins to be deposited in designated cytosolic compartments known as 'aggresomes'. The roles of aggresomes as cellular quality control centers, and the cellular origin of the deposits contained within these structures, remain to be characterized. Here, we utilized the observation that the prion protein (PrP, also known as PRNP) accumulates in aggresomes following the inhibition of folding chaperones, members of the cyclophilin family, to address these questions. We found that misfolded PrP molecules must pass through the endoplasmic reticulum (ER) in order to be deposited in aggresomes, that the Golgi plays no role in this process and that cytosolic PrP species are not deposited in pre-existing aggresomes. Prior to their deposition in the aggresome, PrP molecules lose the ER localization signal and have to acquire a GPI anchor. Our discoveries indicate that PrP aggresomes are cytosolic overflow deposition centers for the ER quality control mechanisms and highlight the importance of these structures for the maintenance of protein homeostasis within the ER.
Collapse
Affiliation(s)
- Tatyana Dubnikov
- Biochemistry and Molecular Biology, the Institute for Medical Research Israel - Canada (IMRIC), the School of Medicine of the Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Tziona Ben-Gedalya
- Biochemistry and Molecular Biology, the Institute for Medical Research Israel - Canada (IMRIC), the School of Medicine of the Hebrew University of Jerusalem, Jerusalem 91120, Israel Department of Obstetrics and Gynecology, Hadassah University Hospital, Ein Kerem, Jerusalem, 91120, Israel
| | - Robert Reiner
- Biochemistry and Molecular Biology, the Institute for Medical Research Israel - Canada (IMRIC), the School of Medicine of the Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Dominic Hoepfner
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel 4056, Switzerland
| | - Wayne A Cabral
- Bone and Extracellular Matrix Branch, NICHD, NIH, Bethesda, MD 20892, USA
| | - Joan C Marini
- Bone and Extracellular Matrix Branch, NICHD, NIH, Bethesda, MD 20892, USA
| | - Ehud Cohen
- Biochemistry and Molecular Biology, the Institute for Medical Research Israel - Canada (IMRIC), the School of Medicine of the Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
31
|
Pisano M, Palomba A, Tanca A, Pagnozzi D, Uzzau S, Addis MF, Dettori MA, Fabbri D, Palmieri G, Rozzo C. Protein expression changes induced in a malignant melanoma cell line by the curcumin analogue compound D6. BMC Cancer 2016; 16:317. [PMID: 27192978 PMCID: PMC4870815 DOI: 10.1186/s12885-016-2362-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/13/2016] [Indexed: 01/09/2023] Open
Abstract
Background We have previously demonstrated that the hydroxylated biphenyl compound D6 (3E,3′E)-4,4′-(5,5′,6,6′-tetramethoxy-[1,1′-biphenyl]-3,3′-diyl)bis(but-3-en-2-one), a structural analogue of curcumin, exerts a strong antitumor activity on melanoma cells both in vitro and in vivo. Although the mechanism of action of D6 is yet to be clarified, this compound is thought to inhibit cancer cell growth by arresting the cell cycle in G2/M phase, and to induce apoptosis through the mitochondrial intrinsic pathway. To investigate the changes in protein expression induced by exposure of melanoma cells to D6, a differential proteomic study was carried out on D6-treated and untreated primary melanoma LB24Dagi cells. Methods Proteins were fractionated by SDS-PAGE and subjected to in gel digestion. The peptide mixtures were analyzed by liquid chromatography coupled with tandem mass spectrometry. Proteins were identified and quantified using database search and spectral counting. Proteomic data were finally uploaded into the Ingenuity Pathway Analysis software to find significantly modulated networks and pathways. Results Analysis of the differentially expressed protein profiles revealed the activation of a strong cellular stress response, with overexpression of several HSPs and stimulation of ubiquitin-proteasome pathways. These were accompanied by a decrease of protein synthesis, evidenced by downregulation of proteins involved in mRNA processing and translation. These findings are consistent with our previous results on gene expression profiling in melanoma cells treated with D6. Conclusions Our findings confirm that the curcumin analogue D6 triggers a strong stress response in melanoma cells, turning down majority of cell functions and finally driving cells to apoptosis. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2362-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marina Pisano
- Institute of Biomolecular Chemistry, National Research Council of Italy, Traversa la Crucca, 3, 07100, Sassari, Italy
| | - Antonio Palomba
- Proteomics Laboratory, Porto Conte Ricerche, Tramariglio, Alghero, Italy.,Biosistema Scrl, Sassari, Italy
| | - Alessandro Tanca
- Proteomics Laboratory, Porto Conte Ricerche, Tramariglio, Alghero, Italy
| | - Daniela Pagnozzi
- Proteomics Laboratory, Porto Conte Ricerche, Tramariglio, Alghero, Italy
| | - Sergio Uzzau
- Proteomics Laboratory, Porto Conte Ricerche, Tramariglio, Alghero, Italy
| | | | - Maria Antonietta Dettori
- Institute of Biomolecular Chemistry, National Research Council of Italy, Traversa la Crucca, 3, 07100, Sassari, Italy
| | - Davide Fabbri
- Institute of Biomolecular Chemistry, National Research Council of Italy, Traversa la Crucca, 3, 07100, Sassari, Italy
| | - Giuseppe Palmieri
- Institute of Biomolecular Chemistry, National Research Council of Italy, Traversa la Crucca, 3, 07100, Sassari, Italy
| | - Carla Rozzo
- Institute of Biomolecular Chemistry, National Research Council of Italy, Traversa la Crucca, 3, 07100, Sassari, Italy.
| |
Collapse
|
32
|
Folda A, Citta A, Scalcon V, Calì T, Zonta F, Scutari G, Bindoli A, Rigobello MP. Mitochondrial Thioredoxin System as a Modulator of Cyclophilin D Redox State. Sci Rep 2016; 6:23071. [PMID: 26975474 PMCID: PMC4791683 DOI: 10.1038/srep23071] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/25/2016] [Indexed: 01/25/2023] Open
Abstract
The mitochondrial thioredoxin system (NADPH, thioredoxin reductase, thioredoxin) is a major redox regulator. Here we have investigated the redox correlation between this system and the mitochondrial enzyme cyclophilin D. The peptidyl prolyl cis-trans isomerase activity of cyclophilin D was stimulated by the thioredoxin system, while it was decreased by cyclosporin A and the thioredoxin reductase inhibitor auranofin. The redox state of cyclophilin D, thioredoxin 1 and 2 and peroxiredoxin 3 was measured in isolated rat heart mitochondria and in tumor cell lines (CEM-R and HeLa) by redox Western blot analysis upon inhibition of thioredoxin reductase with auranofin, arsenic trioxide, 1-chloro-2,4-dinitrobenzene or after treatment with hydrogen peroxide. A concomitant oxidation of thioredoxin, peroxiredoxin and cyclophilin D was observed, suggesting a redox communication between the thioredoxin system and cyclophilin. This correlation was further confirmed by i) co-immunoprecipitation assay of cyclophilin D with thioredoxin 2 and peroxiredoxin 3, ii) molecular modeling and iii) depleting thioredoxin reductase by siRNA. We conclude that the mitochondrial thioredoxin system controls the redox state of cyclophilin D which, in turn, may act as a regulator of several processes including ROS production and pro-apoptotic factors release.
Collapse
Affiliation(s)
- Alessandra Folda
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Anna Citta
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Valeria Scalcon
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Tito Calì
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Francesco Zonta
- Shangai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, No. 99 Haike Road, Pudong, Shanghai 201210, China
| | - Guido Scutari
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Alberto Bindoli
- Institute of Neuroscience (CNR), viale G. Colombo 3, 35131 Padova, Italy
| | - Maria Pia Rigobello
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35131 Padova, Italy
| |
Collapse
|
33
|
Chapman DC, Stocki P, Williams DB. Cyclophilin C Participates in the US2-Mediated Degradation of Major Histocompatibility Complex Class I Molecules. PLoS One 2015; 10:e0145458. [PMID: 26691022 PMCID: PMC4686535 DOI: 10.1371/journal.pone.0145458] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 12/03/2015] [Indexed: 02/07/2023] Open
Abstract
Human cytomegalovirus uses a variety of mechanisms to evade immune recognition through major histocompatibility complex class I molecules. One mechanism mediated by the immunoevasin protein US2 causes rapid disposal of newly synthesized class I molecules by the endoplasmic reticulum-associated degradation pathway. Although several components of this degradation pathway have been identified, there are still questions concerning how US2 targets class I molecules for degradation. In this study we identify cyclophilin C, a peptidyl prolyl isomerase of the endoplasmic reticulum, as a component of US2-mediated immune evasion. Cyclophilin C could be co-isolated with US2 and with the class I molecule HLA-A2. Furthermore, it was required at a particular expression level since depletion or overexpression of cyclophilin C impaired the degradation of class I molecules. To better characterize the involvement of cyclophilin C in class I degradation, we used LC-MS/MS to detect US2-interacting proteins that were influenced by cyclophilin C expression levels. We identified malectin, PDIA6, and TMEM33 as proteins that increased in association with US2 upon cyclophilin C knockdown. In subsequent validation all were shown to play a functional role in US2 degradation of class I molecules. This was specific to US2 rather than general ER-associated degradation since depletion of these proteins did not impede the degradation of a misfolded substrate, the null Hong Kong variant of α1-antitrypsin.
Collapse
Affiliation(s)
- Daniel C. Chapman
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Pawel Stocki
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - David B. Williams
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
34
|
Delaunay-Moisan A, Appenzeller-Herzog C. The antioxidant machinery of the endoplasmic reticulum: Protection and signaling. Free Radic Biol Med 2015; 83:341-51. [PMID: 25744411 DOI: 10.1016/j.freeradbiomed.2015.02.019] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/20/2015] [Accepted: 02/22/2015] [Indexed: 12/16/2022]
Abstract
Cellular metabolism is inherently linked to the production of oxidizing by-products, including reactive oxygen species (ROS) hydrogen peroxide (H2O2). When present in excess, H2O2 can damage cellular biomolecules, but when produced in coordinated fashion, it typically serves as a mobile signaling messenger. It is therefore not surprising that cell health critically relies on both low-molecular-weight and enzymatic antioxidant components, which protect from ROS-mediated damage and shape the propagation and duration of ROS signals. This review focuses on H2O2-antioxidant cross talk in the endoplasmic reticulum (ER), which is intimately linked to the process of oxidative protein folding. ER-resident or ER-regulated sources of H2O2 and other ROS, which are subgrouped into constitutive and stimulated sources, are discussed and set into context with the diverse antioxidant mechanisms in the organelle. These include two types of peroxide-reducing enzymes, a high concentration of glutathione derived from the cytosol, and feedback-regulated thiol-disulfide switches, which negatively control the major ER oxidase ER oxidoreductin-1. Finally, new evidence highlighting emerging principles of H2O2-based cues at the ER will likely set a basis for establishing ER redox processes as a major line of future signaling research. A fundamental problem that remains to be solved is the specific, quantitative, time resolved, and targeted detection of H2O2 in the ER and in specialized ER subdomains.
Collapse
Affiliation(s)
- Agnès Delaunay-Moisan
- Laboratoire Stress Oxydants et Cancer, CEA-Saclay, Service de Biologie Intégrative et de Génétique Moléculaire, Institut de Biologie et de Technologie de Saclay, Commissariat à l׳Energie Atomique et aux Energies Alternatives, F-91191 Gif Sur Yvette, France/Institute for Integrative Biology of the Cell (I2BC), Avenue de la Terrasse, 91198 Gif sur Yvette Cedex, France.
| | | |
Collapse
|
35
|
Tannous A, Pisoni GB, Hebert DN, Molinari M. N-linked sugar-regulated protein folding and quality control in the ER. Semin Cell Dev Biol 2015; 41:79-89. [PMID: 25534658 PMCID: PMC4474783 DOI: 10.1016/j.semcdb.2014.12.001] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/02/2014] [Indexed: 11/18/2022]
Abstract
Asparagine-linked glycans (N-glycans) are displayed on the majority of proteins synthesized in the endoplasmic reticulum (ER). Removal of the outermost glucose residue recruits the lectin chaperone malectin possibly involved in a first triage of defective polypeptides. Removal of a second glucose promotes engagement of folding and quality control machineries built around the ER lectin chaperones calnexin (CNX) and calreticulin (CRT) and including oxidoreductases and peptidyl-prolyl isomerases. Deprivation of the last glucose residue dictates the release of N-glycosylated polypeptides from the lectin chaperones. Correctly folded proteins are authorized to leave the ER. Non-native polypeptides are recognized by the ER quality control key player UDP-glucose glycoprotein glucosyltransferase 1 (UGT1), re-glucosylated and re-addressed to the CNX/CRT chaperone binding cycle to provide additional opportunity for the protein to fold in the ER. Failure to attain the native structure determines the selection of the misfolded polypeptides for proteasome-mediated degradation.
Collapse
Affiliation(s)
- Abla Tannous
- Department of Biochemistry and Molecular Biology, Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA 01003, USA
| | | | - Daniel N Hebert
- Department of Biochemistry and Molecular Biology, Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA 01003, USA.
| | - Maurizio Molinari
- Università della Svizzera italiana, CH-6900 Lugano, Switzerland; Institute for Research in Biomedicine, Protein Folding and Quality Control, CH-6500 Bellinzona, Switzerland; Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
36
|
Ishikawa Y, Boudko S, Bächinger HP. Ziploc-ing the structure: Triple helix formation is coordinated by rough endoplasmic reticulum resident PPIases. Biochim Biophys Acta Gen Subj 2015; 1850:1983-93. [PMID: 25583561 DOI: 10.1016/j.bbagen.2014.12.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/26/2014] [Accepted: 12/29/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND Protein folding is crucial for proteins' specific functions and is facilitated by various types of enzymes and molecular chaperones. The peptidyl prolyl cis/trans isomerases (PPIase) are one of these families of enzymes. They ubiquitously exist inside the cell and there are eight PPIases in the rough endoplasmic reticulum (rER), a compartment where the folding of most secreted proteins occurs. SCOPE OF REVIEW We review the functional and structural aspects of individual rER resident PPIases. Furthermore, we specifically discuss the role of these PPIases during collagen biosynthesis, since collagen is the most abundant protein in humans, is synthesized in the rER, and contains a proportionally high number of proline residues. MAJOR CONCLUSIONS The rER resident PPIases recognize different sets of substrates and facilitate their folding. Although they are clearly catalysts for protein folding, they also have more broad and multifaceted functions. We propose that PPIases coordinate collagen biosynthesis in the rER. GENERAL SIGNIFICANCE This review expands our understanding of collagen biosynthesis by explaining the influence of novel indirect mechanisms of regulating folding and this is also explored for PPIases. We also suggest future directions of research to obtain a better understanding of collagen biosynthesis and functions of PPIases in the rER. This article is part of a Special Issue entitled Proline-directed Foldases: Cell Signaling Catalysts and Drug Targets.
Collapse
Affiliation(s)
- Yoshihiro Ishikawa
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR 97239, USA; Shriners Hospital for Children, Research Department, Portland, OR 97239, USA
| | - Sergei Boudko
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR 97239, USA; Shriners Hospital for Children, Research Department, Portland, OR 97239, USA
| | - Hans Peter Bächinger
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR 97239, USA; Shriners Hospital for Children, Research Department, Portland, OR 97239, USA.
| |
Collapse
|
37
|
Bukrinsky M. Extracellular cyclophilins in health and disease. Biochim Biophys Acta Gen Subj 2014; 1850:2087-95. [PMID: 25445705 PMCID: PMC4436085 DOI: 10.1016/j.bbagen.2014.11.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/11/2014] [Accepted: 11/14/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Extracellular cyclophilins (eCyPs) are pro-inflammatory factors implicated in pathogenesis of a number of inflammatory diseases. Most pathogenic activities of eCyPs are related to their chemotactic action towards leukocytes, which is mediated by eCyP receptor on target cells, CD147, and involves peptidyl-prolyl cis-trans isomerase activity of cyclophilins. This activity is inhibited by cyclosporine A (CsA) and non-immunosuppressive derivatives of this drug. Accumulating evidence for the role of eCyPs in disease pathogenesis stimulated research on the mechanisms of eCyP-initiated events, resulting in identification of multiple signaling pathways, characterization of a variety of effector molecules released from eCyP-treated cells, and synthesis of CsA derivatives specifically blocking eCyPs. However, a number of important questions related to the mode of action of eCyPs remain unanswered. SCOPE OF REVIEW In this article, we integrate available information on release and function of extracellular cyclophilins into a unified model, focusing on outstanding issues that need to be clarified. MAJOR CONCLUSIONS Extracellular cyclophilins are critical players in pathogenesis of a number of inflammatory diseases. Their mechanism of action involves interaction with the receptor, CD147, and initiation of a poorly characterized signal transduction process culminating in chemotaxis and production of pro-inflammatory factors. GENERAL SIGNIFICANCE Extracellular cyclophilins present an attractive target for therapeutic interventions that can be used to alleviate symptoms and consequences of acute and chronic inflammation. This article is part of a Special Issue entitled Proline-directed Foldases: Cell Signaling Catalysts and Drug Targets.
Collapse
Affiliation(s)
- Michael Bukrinsky
- George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| |
Collapse
|