1
|
Yuki S, Sasaki S, Yamamoto Y, Murakami F, Sakata K, Araki I. Evolution of the Cdk4/6-Cdkn2 system in invertebrates. Genes Cells 2024; 29:1037-1051. [PMID: 39380239 PMCID: PMC11555623 DOI: 10.1111/gtc.13165] [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: 05/07/2024] [Revised: 08/15/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
The cell cycle is driven by cyclin-dependent kinases (Cdks). The decision whether the cell cycle proceeds is made during G1 phase, when Cdk4/6 functions. Cyclin-dependent kinase inhibitor 2 (Cdkn2) is a specific inhibitor of Cdk4/6, and their interaction depends on D84 in Cdkn2 and R24/31 in Cdk4/6. This knowledge is based mainly on studies in mammalian cells. Here, we comprehensively analyzed Cdk4/6 and Cdkn2 in invertebrates and found that Cdk4/6 was present in most of the investigated phyla, but the distribution of Cdkn2 was rather uneven among and within the phyla. The positive charge of R24/R31 in Cdk4/6 was conserved in all analyzed species in phyla with Cdkn2. The presence of Cdkn2 and the conservation of the positive charge were statistically correlated. We also found that Cdkn2 has been tightly linked to Fas associated factor 1 (Faf1) during evolution. We discuss potential interactions between Cdkn2 and Cdk4/6 in evolution and the possible cause of the strong conservation of the microsynteny.
Collapse
Affiliation(s)
- Shiori Yuki
- Graduate School of Arts and Sciences, Iwate UniversityMoriokaJapan
| | - Shunsuke Sasaki
- Faculty of Science and Engineering, Iwate UniversityMoriokaJapan
| | - Yuta Yamamoto
- Faculty of Science and Engineering, Iwate UniversityMoriokaJapan
| | - Fumika Murakami
- Graduate School of Arts and Sciences, Iwate UniversityMoriokaJapan
| | - Kazumi Sakata
- Graduate School of Arts and Sciences, Iwate UniversityMoriokaJapan
- Faculty of Science and Engineering, Iwate UniversityMoriokaJapan
| | - Isato Araki
- Graduate School of Arts and Sciences, Iwate UniversityMoriokaJapan
- Faculty of Science and Engineering, Iwate UniversityMoriokaJapan
| |
Collapse
|
2
|
Tanaka LY, Kumar S, Gutierre LF, Magnun C, Kajihara D, Kang DW, Laurindo FRM, Jo H. Disturbed flow regulates protein disulfide isomerase A1 expression via microRNA-204. Front Physiol 2024; 15:1327794. [PMID: 38638277 PMCID: PMC11024637 DOI: 10.3389/fphys.2024.1327794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/13/2024] [Indexed: 04/20/2024] Open
Abstract
Redox processes can modulate vascular pathophysiology. The endoplasmic reticulum redox chaperone protein disulfide isomerase A1 (PDIA1) is overexpressed during vascular proliferative diseases, regulating thrombus formation, endoplasmic reticulum stress adaptation, and structural remodeling. However, both protective and deleterious vascular effects have been reported for PDIA1, depending on the cell type and underlying vascular condition. Further understanding of this question is hampered by the poorly studied mechanisms underlying PDIA1 expression regulation. Here, we showed that PDIA1 mRNA and protein levels were upregulated (average 5-fold) in the intima and media/adventitia following partial carotid ligation (PCL). Our search identified that miR-204-5p and miR-211-5p (miR-204/211), two broadly conserved miRNAs, share PDIA1 as a potential target. MiR-204/211 was downregulated in vascular layers following PCL. In isolated endothelial cells, gain-of-function experiments of miR-204 with miR mimic decreased PDIA1 mRNA while having negligible effects on markers of endothelial activation/stress response. Similar effects were observed in vascular smooth muscle cells (VSMCs). Furthermore, PDIA1 downregulation by miR-204 decreased levels of the VSMC contractile differentiation markers. In addition, PDIA1 overexpression prevented VSMC dedifferentiation by miR-204. Collectively, we report a new mechanism for PDIA1 regulation through miR-204 and identify its relevance in a model of vascular disease playing a role in VSMC differentiation. This mechanism may be regulated in distinct stages of atherosclerosis and provide a potential therapeutic target.
Collapse
Affiliation(s)
- Leonardo Y. Tanaka
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Lucas F. Gutierre
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Celso Magnun
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Daniela Kajihara
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Dong-Won Kang
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Francisco R. M. Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
3
|
Guan IA, Liu JST, Sawyer RC, Li X, Jiao W, Jiramongkol Y, White MD, Hagimola L, Passam FH, Tran DP, Liu X, Schoenwaelder SM, Jackson SP, Payne RJ, Liu X. Integrating Phenotypic and Chemoproteomic Approaches to Identify Covalent Targets of Dietary Electrophiles in Platelets. ACS CENTRAL SCIENCE 2024; 10:344-357. [PMID: 38435523 PMCID: PMC10906253 DOI: 10.1021/acscentsci.3c00822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 03/05/2024]
Abstract
A large variety of dietary phytochemicals has been shown to improve thrombosis and stroke outcomes in preclinical studies. Many of these compounds feature electrophilic functionalities that potentially undergo covalent addition to the sulfhydryl side chain of cysteine residues within proteins. However, the impact of such covalent modifications on the platelet activity and function remains unclear. This study explores the irreversible engagement of 23 electrophilic phytochemicals with platelets, unveiling the unique antiplatelet selectivity of sulforaphane (SFN). SFN impairs platelet responses to adenosine diphosphate (ADP) and a thromboxane A2 receptor agonist while not affecting thrombin and collagen-related peptide activation. It also substantially reduces platelet thrombus formation under arterial flow conditions. Using an alkyne-integrated probe, protein disulfide isomerase A6 (PDIA6) was identified as a rapid kinetic responder to SFN. Mechanistic profiling studies revealed SFN's nuanced modulation of PDIA6 activity and substrate specificity. In an electrolytic injury model of thrombosis, SFN enhanced the thrombolytic activity of recombinant tissue plasminogen activator (rtPA) without increasing blood loss. Our results serve as a catalyst for further investigations into the preventive and therapeutic mechanisms of dietary antiplatelets, aiming to enhance the clot-busting power of rtPA, currently the only approved therapeutic for stroke recanalization that has significant limitations.
Collapse
Affiliation(s)
- Ivy A. Guan
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
| | - Joanna S. T. Liu
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
- School
of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Renata C. Sawyer
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
| | - Xiang Li
- Department
of Medicine, Washington University in St.
Louis, St. Louis, Missouri 63110, United States
- McDonnell
Genome Institute, Washington University
in St. Louis, St. Louis, Missouri 63108, United States
| | - Wanting Jiao
- Ferrier Research
Institute, Victoria University of Wellington, Wellington 6140, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1142, New Zealand
| | - Yannasittha Jiramongkol
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
- Charles
Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Mark D. White
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
| | - Lejla Hagimola
- School
of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Freda H. Passam
- School
of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Denise P. Tran
- Sydney
Mass Spectrometry, The University of Sydney, Camperdown, New South Wales 2006, Australia
| | - Xiaoming Liu
- School
of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Simone M. Schoenwaelder
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
- School
of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Shaun P. Jackson
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
- Charles
Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Richard J. Payne
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
- Australian
Research Council Centre of Excellence for Innovations in Peptide and
Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Xuyu Liu
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
| |
Collapse
|
4
|
Trostchansky A, Alarcon M. An Overview of Two Old Friends Associated with Platelet Redox Signaling, the Protein Disulfide Isomerase and NADPH Oxidase. Biomolecules 2023; 13:biom13050848. [PMID: 37238717 DOI: 10.3390/biom13050848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 12/30/2022] [Accepted: 01/18/2023] [Indexed: 05/28/2023] Open
Abstract
Oxidative stress participates at the baseline of different non-communicable pathologies such as cardiovascular diseases. Excessive formation of reactive oxygen species (ROS), above the signaling levels necessary for the correct function of organelles and cells, may contribute to the non-desired effects of oxidative stress. Platelets play a relevant role in arterial thrombosis, by aggregation triggered by different agonists, where excessive ROS formation induces mitochondrial dysfunction and stimulate platelet activation and aggregation. Platelet is both a source and a target of ROS, thus we aim to analyze both the platelet enzymes responsible for ROS generation and their involvement in intracellular signal transduction pathways. Among the proteins involved in these processes are Protein Disulphide Isomerase (PDI) and NADPH oxidase (NOX) isoforms. By using bioinformatic tools and information from available databases, a complete bioinformatic analysis of the role and interactions of PDI and NOX in platelets, as well as the signal transduction pathways involved in their effects was performed. We focused the study on analyzing whether these proteins collaborate to control platelet function. The data presented in the current manuscript support the role that PDI and NOX play on activation pathways necessary for platelet activation and aggregation, as well as on the platelet signaling imbalance produced by ROS production. Our data could be used to design specific enzyme inhibitors or a dual inhibition for these enzymes with an antiplatelet effect to design promising treatments for diseases involving platelet dysfunction.
Collapse
Affiliation(s)
- Andrés Trostchansky
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Marcelo Alarcon
- Thrombosis Research Center, Universidad de Talca, Talca 3460000, Chile
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| |
Collapse
|
5
|
Functions and mechanisms of protein disulfide isomerase family in cancer emergence. Cell Biosci 2022; 12:129. [PMID: 35965326 PMCID: PMC9375924 DOI: 10.1186/s13578-022-00868-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
The endoplasmic reticulum (ER) is a multi-layered organelle that is essential for the synthesis, folding, and structural maturation of almost one-third of the cellular proteome. It houses several resident proteins for these functions including the 21 members of the protein disulfide isomerase (PDI) family. The signature of proteins belonging to this family is the presence of the thioredoxin domain which mediates the formation, and rearrangement of disulfide bonds of substrate proteins in the ER. This process is crucial not only for the proper folding of ER substrates but also for maintaining a balanced ER proteostasis. The inclusion of new PDI members with a wide variety of structural determinants, size and enzymatic activity has brought additional epitomes of how PDI functions. Notably, some of them do not carry the thioredoxin domain and others have roles outside the ER. This also reflects that PDIs may have specialized functions and their functions are not limited within the ER. Large-scale expression datasets of human clinical samples have identified that the expression of PDI members is elevated in pathophysiological states like cancer. Subsequent functional interrogations using structural, molecular, cellular, and animal models suggest that some PDI members support the survival, progression, and metastasis of several cancer types. Herein, we review recent research advances on PDIs, vis-à-vis their expression, functions, and molecular mechanisms in supporting cancer growth with special emphasis on the anterior gradient (AGR) subfamily. Last, we posit the relevance and therapeutic strategies in targeting the PDIs in cancer.
Collapse
|
6
|
Abstract
Significance: Since protein disulfide isomerase (PDI) was first described in 1963, researchers have shown conclusively that PDI and sibling proteins are quintessential for thrombus formation. PDI, endoplasmic reticulum protein (ERp)5, ERp57, and ERp72 are released from platelets and vascular cells and interact with integrin αIIbβ3 on the outer surface of platelets. Recent Advances: At the cell surface they influence protein folding and function, propagating thrombosis and maintaining hemostasis. TMX1, which is a transmembrane thiol isomerase, is the first family member shown to negatively regulate platelets. Targets of thiol isomerases have been identified, including integrin α2β1, Von Willebrand Factor, GpIbα, nicotinamide adenine dinucleotide phosphate oxidase (Nox)-1, Nox-2, and tissue factor, all of which are pro-thrombotic, and several of which are on the cell surface. In spite of this, PDI can paradoxically catalyze the delivery of nitric oxide to platelets, which decrease thrombus formation. Critical Issues: Although the overall effect of PDI is to positively regulate platelet activation, it is still unclear how thiol isomerases function in pro-thrombotic states, such as obesity, diabetes, and cancer. In parallel, there has been a surge in the development of novel thiol isomerase inhibitors, which display selectivity, potency and modulate thrombosis and hemostasis. The availability of selective thiol isomerase inhibitors has culminated in clinical trials, with promising outcomes for the prevention of cancer-associated thrombosis. Future Directions: Altogether, thiol isomerases are perceived as an orchestrating force that regulates thrombus development. In the current review, we will explore the history of PDI in cardiovascular biology, detail known mechanisms of action, and summarize known thiol isomerase inhibitors.
Collapse
Affiliation(s)
- Renato Simões Gaspar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
7
|
Fernandes DC, Wosniak J, Gonçalves RC, Tanaka LY, Fernandes CG, Zanatta DB, de Mattos ABM, Strauss BE, Laurindo FRM. PDIA1 acts as master organizer of NOX1/NOX4 balance and phenotype response in vascular smooth muscle. Free Radic Biol Med 2021; 162:603-614. [PMID: 33227407 DOI: 10.1016/j.freeradbiomed.2020.11.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/29/2020] [Accepted: 11/17/2020] [Indexed: 02/05/2023]
Abstract
Changes in vascular smooth muscle cell (VSMC) phenotype underlie disease pathophysiology and are strongly regulated by NOX NADPH oxidases, with NOX1 favoring synthetic proliferative phenotype and NOX4 supporting differentiation. Growth factor-triggered NOX1 expression/activity strictly depends on the chaperone oxidoreductase protein disulfide isomerase-A1 (PDIA1). Intracellular PDIA1 is required for VSMC migration and cytoskeleton organization, while extracellular PDIA1 fine-tunes cytoskeletal mechanoadaptation and vascular remodeling. We hypothesized that PDIA1 orchestrates NOX1/NOX4 balance and VSMC phenotype. Using an inducible PDIA1 overexpression model in VSMC, we showed that early PDIA1 overexpression (for 24-48 h) increased NOX1 expression, hydrogen peroxide steady-state levels and spontaneous VSMC migration distances. Sustained PDIA1 overexpression for 72 h and 96 h supported high NOX1 levels while also increasing NOX4 expression and, remarkably, switched VSMC phenotype to differentiation. Differentiation was preceded by increased nuclear myocardin and serum response factor-response element activation, with no change in cell viability. Both NOX1 and hydrogen peroxide were necessary for later PDIA1-induced VSMC differentiation. In primary VSMC, PDIA1 knockdown decreased nuclear myocardin and increased the proliferating cell nuclear antigen expression. Newly-developed PDIA1-overexpressing mice (TgPDIA1) exhibited normal general and cardiovascular baseline phenotypes. However, in TgPDIA1 carotids, NOX1 was decreased while NOX4 and calponin expressions were enhanced, indicating overdifferentiation vs. normal carotids. Moreover, in a rabbit overdistension injury model during late vascular repair, PDIA1 silencing impaired VSMC redifferentiation and NOX1/NOX4 balance. Our results suggest a model in which PDIA1 acts as an upstream organizer of NOX1/NOX4 balance and related VSMC phenotype, accounting for baseline differentiation setpoint.
Collapse
Affiliation(s)
- Denise C Fernandes
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil.
| | - João Wosniak
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Renata C Gonçalves
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Leonardo Y Tanaka
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Carolina G Fernandes
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Daniela B Zanatta
- Viral Vector Laboratory, Center for Translational Research in Oncology/LIM24, Cancer Institute of Sao Paulo, School of Medicine, Sao Paulo, Brazil
| | - Ana Barbosa M de Mattos
- Laboratory of Genetic and Molecular Cardiology, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Bryan E Strauss
- Viral Vector Laboratory, Center for Translational Research in Oncology/LIM24, Cancer Institute of Sao Paulo, School of Medicine, Sao Paulo, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil.
| |
Collapse
|
8
|
Kajihara D, Hon CC, Abdullah AN, Wosniak J, Moretti AIS, Poloni JF, Bonatto D, Hashimoto K, Carninci P, Laurindo FRM. Analysis of splice variants of the human protein disulfide isomerase (P4HB) gene. BMC Genomics 2020; 21:766. [PMID: 33148170 PMCID: PMC7640458 DOI: 10.1186/s12864-020-07164-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/20/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Protein Disulfide Isomerases are thiol oxidoreductase chaperones from thioredoxin superfamily with crucial roles in endoplasmic reticulum proteostasis, implicated in many diseases. The family prototype PDIA1 is also involved in vascular redox cell signaling. PDIA1 is coded by the P4HB gene. While forced changes in P4HB gene expression promote physiological effects, little is known about endogenous P4HB gene regulation and, in particular, gene modulation by alternative splicing. This study addressed the P4HB splice variant landscape. RESULTS Ten protein coding sequences (Ensembl) of the P4HB gene originating from alternative splicing were characterized. Structural features suggest that except for P4HB-021, other splice variants are unlikely to exert thiol isomerase activity at the endoplasmic reticulum. Extensive analyses using FANTOM5, ENCODE Consortium and GTEx project databases as RNA-seq data sources were performed. These indicated widespread expression but significant variability in the degree of isoform expression among distinct tissues and even among distinct locations of the same cell, e.g., vascular smooth muscle cells from different origins. P4HB-02, P4HB-027 and P4HB-021 were relatively more expressed across each database, the latter particularly in vascular smooth muscle. Expression of such variants was validated by qRT-PCR in some cell types. The most consistently expressed splice variant was P4HB-021 in human mammary artery vascular smooth muscle which, together with canonical P4HB gene, had its expression enhanced by serum starvation. CONCLUSIONS Our study details the splice variant landscape of the P4HB gene, indicating their potential role to diversify the functional reach of this crucial gene. P4HB-021 splice variant deserves further investigation in vascular smooth muscle cells.
Collapse
Affiliation(s)
- Daniela Kajihara
- Vascular Biology Laboratory, LIM-64, Heart Institute (InCor), University of Sao Paulo School of Medicine, Av. Eneas Carvalho Aguiar, 44, Annex 2, 9th floor, Sao Paulo, CEP 05403-000, Brazil.,Laboratory for Transcriptome Technology, Division of Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Chung-Chau Hon
- Laboratory for Genome Information Analysis, Division of Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Aimi Naim Abdullah
- Laboratory for Transcriptome Technology, Division of Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - João Wosniak
- Vascular Biology Laboratory, LIM-64, Heart Institute (InCor), University of Sao Paulo School of Medicine, Av. Eneas Carvalho Aguiar, 44, Annex 2, 9th floor, Sao Paulo, CEP 05403-000, Brazil
| | - Ana Iochabel S Moretti
- Vascular Biology Laboratory, LIM-64, Heart Institute (InCor), University of Sao Paulo School of Medicine, Av. Eneas Carvalho Aguiar, 44, Annex 2, 9th floor, Sao Paulo, CEP 05403-000, Brazil
| | - Joice F Poloni
- Department of Molecular Biology and Biotechnology, Biotechnology Center of the Federal University of Rio Grande do Sul, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Diego Bonatto
- Department of Molecular Biology and Biotechnology, Biotechnology Center of the Federal University of Rio Grande do Sul, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Kosuke Hashimoto
- Laboratory for Transcriptome Technology, Division of Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Laboratory of Computational Biology, Institute for Protein Research, Osaka University, Osaka, 565-0871, Japan
| | - Piero Carninci
- Laboratory for Transcriptome Technology, Division of Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, LIM-64, Heart Institute (InCor), University of Sao Paulo School of Medicine, Av. Eneas Carvalho Aguiar, 44, Annex 2, 9th floor, Sao Paulo, CEP 05403-000, Brazil.
| |
Collapse
|
9
|
He C, Ruan F, Jiang S, Zeng J, Yin H, Liu R, Zhang Y, Huang L, Wang C, Ma S, Zuo Z. Black Phosphorus Quantum Dots Cause Nephrotoxicity in Organoids, Mice, and Human Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2001371. [PMID: 32338439 DOI: 10.1002/smll.202001371] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 05/12/2023]
Abstract
Quantum dots (QDs) have numerous potential applications in lighting, engineering, and biomedicine. QDs are mainly excreted through the kidney due to their ultrasmall sizes; thus, the kidneys are target organs of QD toxicity. Here, an organoid screening platform is established and used to study the nephrotoxicity of QDs. Organoids are templated from monodisperse microfluidic Matrigel droplets and found to be homogeneous in both tissue structure and functional recapitulation within a population and suitable for the quantitative screening of toxic doses. Kidney organoids are proved displaying higher sensitivity than 2D-cultured cell lines. Similar to metal-containing QDs, black phosphorus (BP)-QDs are found to have moderate toxicity in the kidney organoids. The nephrotoxicity of BP-QDs are validated in both mice and human renal tubular epithelial cells. BP-QDs are also found to cause insulin insensitivity and endoplasmic reticulum (ER) stress in the kidney. Furthermore, ER stress-related IRE1α signaling is shown to mediate renal toxicity and insulin insensitivity caused by BP-QDs. In summary, this work demonstrates the use of constructed kidney organoids as 3D high-throughput screening tools to assess nanosafety and further illuminates the effects and molecular mechanisms of BP-QD nephrotoxicity. The findings will hopefully enable improvement of the safety of BP-QD applications.
Collapse
Affiliation(s)
- Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Fengkai Ruan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shengwei Jiang
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, 518055, China
| | - Jie Zeng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Hanying Yin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Rong Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yongxing Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, Fujian, 361102, China
| | - Laiqiang Huang
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, 518055, China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Shaohua Ma
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, 518055, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| |
Collapse
|
10
|
Urate hydroperoxide oxidizes endothelial cell surface protein disulfide isomerase-A1 and impairs adherence. Biochim Biophys Acta Gen Subj 2019; 1864:129481. [PMID: 31734460 DOI: 10.1016/j.bbagen.2019.129481] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/07/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Extracellular surface protein disulfide isomerase-A1 (PDI) is involved in platelet aggregation, thrombus formation and vascular remodeling. PDI performs redox exchange with client proteins and, hence, its oxidation by extracellular molecules might alter protein function and cell response. In this study, we investigated PDI oxidation by urate hydroperoxide, a newly-described oxidant that is generated through uric acid oxidation by peroxidases, with a putative role in vascular inflammation. METHODS Amino acids specificity and kinetics of PDI oxidation by urate hydroperoxide was evaluated by LC-MS/MS and by stopped-flow. Oxidation of cell surface PDI and other thiol-proteins from HUVECs was identified using impermeable alkylating reagents. Oxidation of intracellular GSH and GSSG was evaluated with specific LC-MS/MS techniques. Cell adherence, detachment and viability were assessed using crystal violet staining, cellular microscopy and LDH activity, respectively. RESULTS Urate hydroperoxide specifically oxidized cysteine residues from catalytic sites of recombinant PDI with a rate constant of 6 × 103 M-1 s-1. Incubation of HUVECs with urate hydroperoxide led to oxidation of cell surface PDI and other unidentified cell surface thiol-proteins. Cell adherence to fibronectin coated plates was impaired by urate hydroperoxide, as well as by other oxidants, thiol alkylating agents and PDI inhibitors. Urate hydroperoxide did not affect cell viability but significantly decreased GSH/GSSG ratio. CONCLUSIONS Our results demonstrated that urate hydroperoxide affects thiol-oxidation of PDI and other cell surface proteins, impairing cellular adherence. GENERAL SIGNIFICANCE These findings could contribute to a better understanding of the mechanism by which uric acid affects endothelial cell function and vascular homeostasis.
Collapse
|
11
|
Oliveira PVSD, Garcia-Rosa S, Sachetto ATA, Moretti AIS, Debbas V, De Bessa TC, Silva NT, Pereira ADC, Martins-de-Souza D, Santoro ML, Laurindo FRM. Protein disulfide isomerase plasma levels in healthy humans reveal proteomic signatures involved in contrasting endothelial phenotypes. Redox Biol 2019; 22:101142. [PMID: 30870787 PMCID: PMC6430080 DOI: 10.1016/j.redox.2019.101142] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/22/2019] [Accepted: 02/12/2019] [Indexed: 12/18/2022] Open
Abstract
Redox-related plasma proteins are candidate reporters of protein signatures associated with endothelial structure/function. Thiol-proteins from protein disulfide isomerase (PDI) family are unexplored in this context. Here, we investigate the occurrence and physiological significance of a circulating pool of PDI in healthy humans. We validated an assay for detecting PDI in plasma of healthy individuals. Our results indicate high inter-individual (median = 330 pg/mL) but low intra-individual variability over time and repeated measurements. Remarkably, plasma PDI levels could discriminate between distinct plasma proteome signatures, with PDI-rich (>median) plasma differentially expressing proteins related to cell differentiation, protein processing, housekeeping functions and others, while PDI-poor plasma differentially displayed proteins associated with coagulation, inflammatory responses and immunoactivation. Platelet function was similar among individuals with PDI-rich vs. PDI-poor plasma. Remarkably, such protein signatures closely correlated with endothelial function and phenotype, since cultured endothelial cells incubated with PDI-poor or PDI-rich plasma recapitulated gene expression and secretome patterns in line with their corresponding plasma signatures. Furthermore, such signatures translated into functional responses, with PDI-poor plasma promoting impairment of endothelial adhesion to fibronectin and a disturbed pattern of wound-associated migration and recovery area. Patients with cardiovascular events had lower PDI levels vs. healthy individuals. This is the first study describing PDI levels as reporters of specific plasma proteome signatures directly promoting contrasting endothelial phenotypes and functional responses.
Collapse
Affiliation(s)
- Percíllia Victória Santos de Oliveira
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Sheila Garcia-Rosa
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico, Sao Paulo, Brazil
| | | | - Ana Iochabel Soares Moretti
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Victor Debbas
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Tiphany Coralie De Bessa
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Nathalia Tenguan Silva
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Alexandre da Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico, Sao Paulo, Brazil
| | | | - Francisco Rafael Martins Laurindo
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
12
|
De Bessa TC, Pagano A, Moretti AIS, Oliveira PVS, Mendonça SA, Kovacic H, Laurindo FRM. Subverted regulation of Nox1 NADPH oxidase-dependent oxidant generation by protein disulfide isomerase A1 in colon carcinoma cells with overactivated KRas. Cell Death Dis 2019; 10:143. [PMID: 30760703 PMCID: PMC6374413 DOI: 10.1038/s41419-019-1402-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 12/24/2018] [Accepted: 01/28/2019] [Indexed: 12/11/2022]
Abstract
Protein disulfide isomerases including PDIA1 are implicated in cancer progression, but underlying mechanisms are unclear. PDIA1 is known to support vascular Nox1 NADPH oxidase expression/activation. Since deregulated reactive oxygen species (ROS) production underlies tumor growth, we proposed that PDIA1 is an upstream regulator of tumor-associated ROS. We focused on colorectal cancer (CRC) with distinct KRas activation levels. Analysis of RNAseq databanks and direct validation indicated enhanced PDIA1 expression in CRC with constitutive high (HCT116) vs. moderate (HKE3) and basal (Caco2) Ras activity. PDIA1 supported Nox1-dependent superoxide production in CRC; however, we first reported a dual effect correlated with Ras-level activity: in Caco2 and HKE3 cells, loss-of-function experiments indicate that PDIA1 sustains Nox1-dependent superoxide production, while in HCT116 cells PDIA1 restricted superoxide production, a behavior associated with increased Rac1 expression/activity. Transfection of Rac1G12V active mutant into HKE3 cells induced PDIA1 to become restrictive of Nox1-dependent superoxide, while in HCT116 cells treated with Rac1 inhibitor, PDIA1 became supportive of superoxide. PDIA1 silencing promoted diminished cell proliferation and migration in HKE3, not detectable in HCT116 cells. Screening of cell signaling routes affected by PDIA1 silencing highlighted GSK3β and Stat3. Also, E-cadherin expression after PDIA1 silencing was decreased in HCT116, consistent with PDIA1 support of epithelial-mesenchymal transition. Thus, Ras overactivation switches the pattern of PDIA1-dependent Rac1/Nox1 regulation, so that Ras-induced PDIA1 bypass can directly activate Rac1. PDIA1 may be a crucial regulator of redox-dependent adaptive processes related to cancer progression.
Collapse
Affiliation(s)
- Tiphany Coralie De Bessa
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Faculté de Pharmacie, 27, Boulevard Jean Moulin - 13385 Marseille CEDEX 5-France, Marseille, France
| | - Alessandra Pagano
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Faculté de Pharmacie, 27, Boulevard Jean Moulin - 13385 Marseille CEDEX 5-France, Marseille, France
| | - Ana Iochabel Soares Moretti
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Percillia Victoria Santos Oliveira
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Samir Andrade Mendonça
- Centro de Investigação Translacional em Oncologia do Instituto do Câncer do Estado de São Paulo (Icesp), Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Herve Kovacic
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Faculté de Pharmacie, 27, Boulevard Jean Moulin - 13385 Marseille CEDEX 5-France, Marseille, France.
| | - Francisco Rafael Martins Laurindo
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
13
|
Tanaka LY, Araujo TLS, Rodriguez AI, Ferraz MS, Pelegati VB, Morais MCC, Santos AMD, Cesar CL, Ramos AF, Alencar AM, Laurindo FRM. Peri/epicellular protein disulfide isomerase-A1 acts as an upstream organizer of cytoskeletal mechanoadaptation in vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 2018; 316:H566-H579. [PMID: 30499716 DOI: 10.1152/ajpheart.00379.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although redox processes closely interplay with mechanoresponses to control vascular remodeling, redox pathways coupling mechanostimulation to cellular cytoskeletal organization remain unclear. The peri/epicellular pool of protein disulfide isomerase-A1 (pecPDIA1) supports postinjury vessel remodeling. Using distinct models, we investigated whether pecPDIA1 could work as a redox-dependent organizer of cytoskeletal mechanoresponses. In vascular smooth muscle cells (VSMCs), pecPDIA1 immunoneutralization impaired stress fiber assembly in response to equibiaxial stretch and, under uniaxial stretch, significantly perturbed cell repositioning perpendicularly to stretch orientation. During cyclic stretch, pecPDIA1 supported thiol oxidation of the known mechanosensor β1-integrin and promoted polarized compartmentalization of sulfenylated proteins. Using traction force microscopy, we showed that pecPDIA1 organizes intracellular force distribution. The net contractile moment ratio of platelet-derived growth factor-exposed to basal VSMCs decreased from 0.90 ± 0.09 (IgG-exposed controls) to 0.70 ± 0.08 after pecPDI neutralization ( P < 0.05), together with an enhanced coefficient of variation for distribution of force modules, suggesting increased noise. Moreover, in a single cell model, pecPDIA1 neutralization impaired migration persistence without affecting total distance or velocity, whereas siRNA-mediated total PDIA1 silencing disabled all such variables of VSMC migration. Neither expression nor total activity of the master mechanotransmitter/regulator RhoA was affected by pecPDIA1 neutralization. However, cyclic stretch-induced focal distribution of membrane-bound RhoA was disrupted by pecPDI inhibition, which promoted a nonpolarized pattern of RhoA/caveolin-3 cluster colocalization. Accordingly, FRET biosensors showed that pecPDIA1 supports localized RhoA activity at cell protrusions versus perinuclear regions. Thus, pecPDI acts as a thiol redox-dependent organizer and noise reducer mechanism of cytoskeletal repositioning, oxidant generation, and localized RhoA activation during a variety of VSMC mechanoresponses. NEW & NOTEWORTHY Effects of a peri/epicellular pool of protein disulfide isomerase-A1 (pecPDIA1) during mechanoregulation in vascular smooth muscle cells (VSMCs) were highlighted using approaches such as equibiaxial and uniaxial stretch, random single cell migration, and traction force microscopy. pecPDIA1 regulates organization of the cytoskeleton and minimizes the noise of cell alignment, migration directionality, and persistence. pecPDIA1 mechanisms involve redox control of β1-integrin and localized RhoA activation. pecPDIA1 acts as a novel organizer of mechanoadaptation responses in VSMCs.
Collapse
Affiliation(s)
- Leonardo Y Tanaka
- Vascular Biology Laboratory, Heart Institute, University of São Paulo School of Medicine , São Paulo , Brazil
| | - Thaís L S Araujo
- Vascular Biology Laboratory, Heart Institute, University of São Paulo School of Medicine , São Paulo , Brazil
| | - Andres I Rodriguez
- Vascular Biology Laboratory, Heart Institute, University of São Paulo School of Medicine , São Paulo , Brazil.,Group of Research and Innovation in Vascular Health, Department of Basic Sciences, Faculty of Sciences, University of Bío-Bío , Chillán , Chile
| | - Mariana S Ferraz
- Institute of Physics, University of São Paulo , São Paulo , Brazil
| | - Vitor B Pelegati
- "Gleb Wataghin" Institute of Physics, University of Campinas , Campinas , Brazil
| | - Mauro C C Morais
- Escola de Artes, Ciências e Humanidades e Núcleo de Estudos Interdisciplinares em Sistemas Complexos, Departamento de Radiologia e Oncologia e Centro de Pesquisa Translacional em Oncologia - Instituto do Cancer do Estado São Paulo, Faculdade de Medicina, Universidade de São Paulo , São Paulo , Brazil
| | - Aline M Dos Santos
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas , Campinas , Brazil
| | - Carlos L Cesar
- "Gleb Wataghin" Institute of Physics, University of Campinas , Campinas , Brazil
| | - Alexandre F Ramos
- Escola de Artes, Ciências e Humanidades e Núcleo de Estudos Interdisciplinares em Sistemas Complexos, Departamento de Radiologia e Oncologia e Centro de Pesquisa Translacional em Oncologia - Instituto do Cancer do Estado São Paulo, Faculdade de Medicina, Universidade de São Paulo , São Paulo , Brazil
| | | | - Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute, University of São Paulo School of Medicine , São Paulo , Brazil
| |
Collapse
|
14
|
Ferrero E, Faini AC, Malavasi F. A phylogenetic view of the leukocyte ectonucleotidases. Immunol Lett 2018; 205:51-58. [PMID: 29958894 DOI: 10.1016/j.imlet.2018.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022]
Abstract
The leukocyte ectonucleotidases are a recently defined family included in the last Human Leukocyte Differentiation Antigens Workshop, giving prominence to these membrane proteins whose catalytic activity is expressed outside the cell. Among the most important substrates of the leukocyte ectonucleotidases are extracellular ATP and NAD+ whose transient increases are not immunologically silent but rather perceived as danger signals by the host. Among the host responses to the release of ATP, NAD+ and related small molecules is their breakdown on behalf of a panel of leukocyte ectonucleotidases - CD38, CD39, CD73, CD157, CD203a and CD203c -, whose activities are concatenated to form two nucleotide-catabolizing channels defined as the canonical and non-canonical adenosinergic pathways. Here, after briefly reviewing the structure and function of the proteins involved in these pathwys, we focus on the genes encoding the ectoenzymes of these adenosinergic pathways. The chromosomal localizations of the enzyme-encoding genes yield a first level of information concerning their origins by duplication and modes of regulation. Further information was obtained from phylogenetic analyses that show ectoenzyme orthologs are conserved in major tetrapod species whereas examination of synteny conservation revealed that the chromosomal regions harboring the ADP-ribosyl cyclases on human chromosome 4 and the ENTPDase CD39 on chromosome 10 show striking similarities in gene content consistent with their being paralogous chromosomal regions derived from a vertebrate whole genome duplication. Thus the connections between some of the leukocyte ectoenzymes run deeper than previously imagined.
Collapse
Affiliation(s)
- Enza Ferrero
- Immunogenetics Laboratory, Department of Medical Sciences, University of Torino, Torino, Italy.
| | - Angelo C Faini
- Immunogenetics Laboratory, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Fabio Malavasi
- Immunogenetics Laboratory, Department of Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
15
|
Campos JLO, Doratioto TR, Videira NB, Ribeiro Filho HV, Batista FAH, Fattori J, Indolfo NDC, Nakahira M, Bajgelman MC, Cvoro A, Laurindo FRM, Webb P, Figueira ACM. Protein Disulfide Isomerase Modulates the Activation of Thyroid Hormone Receptors. Front Endocrinol (Lausanne) 2018; 9:784. [PMID: 30671024 PMCID: PMC6331412 DOI: 10.3389/fendo.2018.00784] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 12/12/2018] [Indexed: 12/30/2022] Open
Abstract
Thyroid hormone receptors (TRs) are responsible for mediating thyroid hormone (T3 and T4) actions at a cellular level. They belong to the nuclear receptor (NR) superfamily and execute their main functions inside the cell nuclei as hormone-regulated transcription factors. These receptors also exhibit so-called "non-classic" actions, for which other cellular proteins, apart from coregulators inside nuclei, regulate their activity. Aiming to find alternative pathways of TR modulation, we searched for interacting proteins and found that PDIA1 interacts with TRβ in a yeast two-hybrid screening assay. The functional implications of PDIA1-TR interactions are still unclear; however, our co-immunoprecipitation (co-IP) and fluorescence assay results showed that PDI was able to bind both TR isoforms in vitro. Moreover, T3 appears to have no important role in these interactions in cellular assays, where PDIA1 was able to regulate transcription of TRα and TRβ-mediated genes in different ways depending on the promoter region and on the TR isoform involved. Although PDIA1 appears to act as a coregulator, it binds to a TR surface that does not interfere with coactivator binding. However, the TR:PDIA1 complex affinity and activation are different depending on the TR isoform. Such differences may reflect the structural organization of the PDIA1:TR complex, as shown by models depicting an interaction interface with exposed cysteines from both proteins, suggesting that PDIA1 might modulate TR by its thiol reductase/isomerase activity.
Collapse
Affiliation(s)
- Jessica L. O. Campos
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research Energy and Materials (CNPEM), São Paulo, Brazil
- Graduation Program of Biosciences and Bioactive Products Technology, Institute of Biology, State University of Campinas (Unicamp), São Paulo, Brazil
| | - Tabata R. Doratioto
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research Energy and Materials (CNPEM), São Paulo, Brazil
- Graduation Program of Biosciences and Bioactive Products Technology, Institute of Biology, State University of Campinas (Unicamp), São Paulo, Brazil
| | - Natalia B. Videira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research Energy and Materials (CNPEM), São Paulo, Brazil
- Graduation Program of Biosciences and Bioactive Products Technology, Institute of Biology, State University of Campinas (Unicamp), São Paulo, Brazil
| | - Helder V. Ribeiro Filho
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research Energy and Materials (CNPEM), São Paulo, Brazil
- Graduation Program of Biosciences and Bioactive Products Technology, Institute of Biology, State University of Campinas (Unicamp), São Paulo, Brazil
| | - Fernanda A. H. Batista
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research Energy and Materials (CNPEM), São Paulo, Brazil
| | - Juliana Fattori
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research Energy and Materials (CNPEM), São Paulo, Brazil
| | - Nathalia de C. Indolfo
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research Energy and Materials (CNPEM), São Paulo, Brazil
- Graduation Program of Biosciences and Bioactive Products Technology, Institute of Biology, State University of Campinas (Unicamp), São Paulo, Brazil
| | - Marcel Nakahira
- Institute of Chemistry (IQ), State University of Campinas (Unicamp), São Paulo, Brazil
| | - Marcio C. Bajgelman
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research Energy and Materials (CNPEM), São Paulo, Brazil
| | - Aleksandra Cvoro
- Genomic Medicine, The Methodist Hospital Research Institute, Houston, TX, United States
| | - Francisco R. M. Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Paul Webb
- California Institute for Regenerative Medicine, Oakland, CA, United States
| | - Ana Carolina M. Figueira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research Energy and Materials (CNPEM), São Paulo, Brazil
- *Correspondence: Ana Carolina M. Figueira
| |
Collapse
|