1
|
Nie Q, Yang J, Zhou X, Li N, Zhang J. The Role of Protein Disulfide Isomerase Inhibitors in Cancer Therapy. ChemMedChem 2025; 20:e202400590. [PMID: 39319369 DOI: 10.1002/cmdc.202400590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 09/26/2024]
Abstract
Protein disulfide isomerase (PDI) is a member of the mercaptan isomerase family, primarily located in the endoplasmic reticulum (ER). At least 21 PDI family members have been identified. PDI plays a key role in protein folding, correcting misfolded proteins, and catalyzing disulfide bond formation, rearrangement, and breaking. It also acts as a molecular chaperone. Dysregulation of PDI activity is thus linked to diseases such as cancer, infections, immune disorders, thrombosis, neurodegenerative diseases, and metabolic disorders. In particular, elevated intracellular PDI levels can enhance cancer cell proliferation, metastasis, and invasion, making it a potential cancer marker. Cancer cells require extensive protein synthesis, with disulfide bond formation by PDI being a critical producer. Thus, cancer cells have higher PDI levels than normal cells. Targeting PDI can induce ER stress and activate the Unfolded Protein Response (UPR) pathway, leading to cancer cell apoptosis. This review discusses the structure and function of PDI, PDI inhibitors in cancer therapy, and the limitations of current inhibitors, proposing especially future directions for developing new PDI inhibitors.
Collapse
Affiliation(s)
- Qiuying Nie
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, 730000, China
| | - Junwei Yang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, 730000, China
| | - Xiedong Zhou
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, 730000, China
| | - Na Li
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, 730000, China
| | - Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
2
|
Lu Q, Ding Y, Liu W, Liu S. Viral Infections and the Glutathione Peroxidase Family: Mechanisms of Disease Development. Antioxid Redox Signal 2024. [PMID: 39446976 DOI: 10.1089/ars.2024.0645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Significance: The glutathione peroxidase (GPx) family is recognized for its essential function in maintaining cellular redox balance and countering the overproduction of reactive oxygen species (ROS), a process intricately linked to the progression of various diseases including those spurred by viral infections. The modulation of GPx activity by viruses presents a critical juncture in disease pathogenesis, influencing cellular responses and the trajectory of infection-induced diseases. Recent Advances: Cutting-edge research has unveiled the GPx family's dynamic role in modulating viral pathogenesis. Notably, GPX4's pivotal function in regulating ferroptosis presents a novel avenue for the antiviral therapy. The discovery that selenium, an essential micronutrient for GPx activity, possesses antiviral properties has propelled us toward rethinking traditional treatment modalities. Critical Issues: Deciphering the intricate relationship between viral infections and GPx family members is paramount. Viral invasion can precipitate significant alterations in GPx function, influencing disease outcomes. The multifaceted nature of GPx activity during viral infections suggests that a deeper understanding of these interactions could yield novel insights into disease mechanisms, diagnostics, prognostics, and even chemotherapeutic resistance. Future Directions: This review aims to synthesize current knowledge on the impact of viral infections on GPx activity and expression and identify key advances. By elucidating the mechanisms through which GPx family members intersect with viral pathogenesis, we propose to uncover innovative therapeutic strategies that leverage the antioxidant properties of GPx to combat viral infections. The exploration of GPx as a therapeutic target and biomarker holds promise for the development of next-generation antiviral therapies. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Qingqing Lu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Pathogenic Biology, Qingdao University Medical College, Qingdao, China
| | - Yuan Ding
- Department of Special Examination, Qingdao Women and Children's Hospital, Qingdao University, Qingdao, China
| | - Wen Liu
- Department of Pathogenic Biology, Qingdao University Medical College, Qingdao, China
| | - Shuzhen Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
3
|
Luo Q, Mao J, Li Y, Wang M, Zhang L, Shen Z. Molecular characterization of a novel thioredoxin-related transmembrane protein gene AcTMX3 that plays important roles in antioxidant defence in Arma chinensis diapause. INSECT MOLECULAR BIOLOGY 2024. [PMID: 39440724 DOI: 10.1111/imb.12964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
Protein disulphide isomerase (PDI) possesses disulphide isomerase, oxidoreductase and molecular chaperone activities, and is involved in regulating various physiological processes. However, there are few studies on the function in insect diapause. In this study, we cloned one novel member PDI family (TMX3, thioredoxin-related transmembrane protein 3) in Arma chinensis. The AcTMX3 encodes 426 amino acids that contains a predicted N-terminal signal sequence, a thioredoxin-like domain with the CXXC active site and a potential transmembrane region, which are typical sequence features of TMX3. RT-qPCR results showed that AcTMX3 was mainly expressed in the head under non-diapause conditions, while AcTMX3 was highly expressed in the fat body (central metabolic organ) under diapause conditions. Moreover, temporal expression profile showed that compared with non-diapause conditions, diapause conditions significantly induced AcTMX3 expression, and the expression of AcTMX3 was enhanced at 15°C. Silencing AcTMX3 in A. chinensis significantly inhibited the expression of antioxidant genes (AcTrx2 and AcTrx-like), increased the content of H2O2 and ascorbate and reduced the survival rate of A. chinensis under diapause conditions. Our results suggested that AcTMX3 played an important role in the resistance of A. chinensis to oxidative stress under diapause conditions.
Collapse
Affiliation(s)
- Qiaozhi Luo
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Natural Enemy Insects of Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jianjun Mao
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Natural Enemy Insects of Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yuyan Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Natural Enemy Insects of Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mengqing Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Natural Enemy Insects of Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lisheng Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Natural Enemy Insects of Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Zhongjian Shen
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Natural Enemy Insects of Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
4
|
Ji YW, Wen XY, Tang HP, Jin ZS, Su WT, Zhou L, Xia ZY, Xia ZY, Lei SQ. DJ-1: Potential target for treatment of myocardial ischemia-reperfusion injury. Biomed Pharmacother 2024; 179:117383. [PMID: 39232383 DOI: 10.1016/j.biopha.2024.117383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Ischemic heart disease (IHD) is a significant global health concern, resulting in high rates of mortality and disability among patients. Although coronary blood flow reperfusion is a key treatment for IHD, it often leads to acute myocardial ischemia-reperfusion injury (IRI). Current intervention strategies have limitations in providing adequate protection for the ischemic myocardium. DJ-1, originally known as a Parkinson's disease related protein, is a highly conserved cytoprotective protein. It is involved in enhancing mitochondrial function, scavenging reactive oxygen species (ROS), regulating autophagy, inhibiting apoptosis, modulating anaerobic metabolism, and exerting anti-inflammatory effects. DJ-1 is also required for protective strategies, such as ischemic preconditioning, ischemic postconditioning, remote ischemic preconditioning and pharmacological conditioning. Therefore, DJ-1 emerges as a potential target for the treatment of myocardial IRI. Our comprehensive review delves into its protective mechanisms in myocardial IRI and the structural foundations underlying its functions.
Collapse
Affiliation(s)
- Yan-Wei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin-Yu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - He-Peng Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhen-Shuai Jin
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wa-Ting Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lu Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zheng-Yuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shao-Qing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
5
|
Meschiari G, Minacori M, Fiorini S, Tedesco M, Eufemi M, Altieri F. Analysis of Punicalin and Punicalagin Interaction with PDIA3 and PDIA1. Int J Mol Sci 2024; 25:10531. [PMID: 39408858 PMCID: PMC11476419 DOI: 10.3390/ijms251910531] [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: 08/26/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/19/2024] Open
Abstract
PDIA3 is a pleiotropic protein primarily located in the endoplasmic reticulum where it is involved in protein folding, catalyzing the formation, breakage, and rearrangement of disulfide bonds. PDIA3 is implicated in numerous pathologies such as cancer, inflammation, and neurodegeneration. Although punicalagin has been proven to be a highly promising PDIA3 inhibitor and can be used as target protein in glioblastoma, it does not have sufficient selectivity for PDIA3 and is a quite-large molecule. With the aim of finding punicalagin derivatives with a simplified structure, we selected punicalin, which lacks the hexahydroxy-diphenic acid moiety. Previous docking studies suggest that this part of the molecule is not involved in the binding with PDIA3. In this study we compared the ability of punicalin to bind and inhibit PDIA3 and PDIA1. Tryptophan fluorescence quenching and disulfide reductase activity (using both glutathione and insulin as substrates) were evaluated, demonstrating the ability of punicalin to bind and inhibit PDIA3 even to a lesser extent compared to punicalagin. On the other hand, punicalin showed a very low inhibition activity towards PDIA1, demonstrating a higher selectivity for PDIA3. Protein thermal shift assay evidenced that both proteins can be destabilized by punicalin as well as punicalagin, with PDIA3 much more sensitive. Additionally, punicalin showed a higher change in the thermal stability of PDIA3, with a shift up to 8 °C. This result could explain the presence of PDIA3 aggregates, evidenced by immunofluorescence analysis, that accumulate within treated cells and that are more evident in the presence of punicalin. The results here obtained show punicalin is able to bind both proteins but with a higher selectivity for PDIA3, suggesting the possibility of developing new molecules with a simplified structure that are still able to selectively bind and inhibit PDIA3.
Collapse
Affiliation(s)
- Giorgia Meschiari
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Farmacy and Medicine, Sapienza University of Rome, Pl. A. Moro 5, 00185 Rome, Italy; (G.M.); (S.F.); (M.T.); (M.E.)
| | - Marco Minacori
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Campus “Aurelio Saliceti”, Via R. Balzarini 1, 64100 Teramo, Italy;
| | - Sara Fiorini
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Farmacy and Medicine, Sapienza University of Rome, Pl. A. Moro 5, 00185 Rome, Italy; (G.M.); (S.F.); (M.T.); (M.E.)
| | - Mariassunta Tedesco
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Farmacy and Medicine, Sapienza University of Rome, Pl. A. Moro 5, 00185 Rome, Italy; (G.M.); (S.F.); (M.T.); (M.E.)
| | - Margherita Eufemi
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Farmacy and Medicine, Sapienza University of Rome, Pl. A. Moro 5, 00185 Rome, Italy; (G.M.); (S.F.); (M.T.); (M.E.)
| | - Fabio Altieri
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Farmacy and Medicine, Sapienza University of Rome, Pl. A. Moro 5, 00185 Rome, Italy; (G.M.); (S.F.); (M.T.); (M.E.)
| |
Collapse
|
6
|
Niu Y, Wang N, Xu Q. Development of an Endoplasmic Reticulum Stress-Related Diagnostic Signature in Polycystic Ovary Syndrome. Reprod Sci 2024:10.1007/s43032-024-01619-3. [PMID: 38955938 DOI: 10.1007/s43032-024-01619-3] [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: 12/14/2023] [Accepted: 06/03/2024] [Indexed: 07/04/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder in premenopausal women. This investigation was to elucidate the underlying mechanism of endoplasmic reticulum stress (ERS) activation in granulosa cells, which has been implicated in the etiology of PCOS. Differentially expressed genes (DEGs) between PCOS and control groups were integrated with ERS gene lists from databases to identify DE-ERS genes, and functional analyses were performed. Univariate regression analysis and the LASSO method were used to select diagnostic factors, followed by establishing a DE-ERS gene-based diagnostic model. A nomogram model was further generated to predict the risk of PCOS. The correlation between ERS gene expression and immune cell proportion was assessed. A total of 14 DE-ERS genes associated with "protein processing in endoplasmic reticulum", "ferroptosis", and "glycerophospholipid metabolism" were selected as PCOS-related factors. An eight-DE-ERS genes-based diagnostic model was developed and displayed satisfactory performance in the training (Area under curve (AUC) = 0.983) and validation datasets (AUC = 0.802). High risk of PCOS can be accurately predicted, which might contribute to clinical decision-making. Moreover, EDEM1 expression was significantly positively correlated with naive B cell infiltration, while PDIA6 was negatively correlated with neutrophil proportion (P < 0.001). We identified eight novel molecules and developed an ERS gene-based diagnostic model in PCOS, which might provide novel insight for finding biomarkers and treatment methods.
Collapse
Affiliation(s)
- Yanxin Niu
- Department of Obstetrics and Gynaecology, Jinhua People's Hospital, No.267, Danxi East Road, Jinhua, 321000, Zhejiang, P.R. China
| | - Nan Wang
- Department of Obstetrics and Gynaecology, Jinhua People's Hospital, No.267, Danxi East Road, Jinhua, 321000, Zhejiang, P.R. China
| | - Qiulian Xu
- Department of Obstetrics and Gynaecology, Jinhua People's Hospital, No.267, Danxi East Road, Jinhua, 321000, Zhejiang, P.R. China.
| |
Collapse
|
7
|
Gama Cavalcante AL, Dari DN, Izaias da Silva Aires F, Carlos de Castro E, Moreira Dos Santos K, Sousa Dos Santos JC. Advancements in enzyme immobilization on magnetic nanomaterials: toward sustainable industrial applications. RSC Adv 2024; 14:17946-17988. [PMID: 38841394 PMCID: PMC11151160 DOI: 10.1039/d4ra02939a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024] Open
Abstract
Enzymes are widely used in biofuels, food, and pharmaceuticals. The immobilization of enzymes on solid supports, particularly magnetic nanomaterials, enhances their stability and catalytic activity. Magnetic nanomaterials are chosen for their versatility, large surface area, and superparamagnetic properties, which allow for easy separation and reuse in industrial processes. Researchers focus on the synthesis of appropriate nanomaterials tailored for specific purposes. Immobilization protocols are predefined and adapted to both enzymes and support requirements for optimal efficiency. This review provides a detailed exploration of the application of magnetic nanomaterials in enzyme immobilization protocols. It covers methods, challenges, advantages, and future perspectives, starting with general aspects of magnetic nanomaterials, their synthesis, and applications as matrices for solid enzyme stabilization. The discussion then delves into existing enzymatic immobilization methods on magnetic nanomaterials, highlighting advantages, challenges, and potential applications. Further sections explore the industrial use of various enzymes immobilized on these materials, the development of enzyme-based bioreactors, and prospects for these biocatalysts. In summary, this review provides a concise comparison of the use of magnetic nanomaterials for enzyme stabilization, highlighting potential industrial applications and contributing to manufacturing optimization.
Collapse
Affiliation(s)
- Antônio Luthierre Gama Cavalcante
- Departamento de Química Orgânica e Inorgânica, Centro de Ciências, Universidade Federal do Ceará Campus Pici Fortaleza CEP 60455760 CE Brazil
| | - Dayana Nascimento Dari
- Instituto de Engenharias e Desenvolvimento Sustentável, Universidade da Integração Internacional da Lusofonia Afro-Brasileira Campus das Auroras Redenção CEP 62790970 CE Brazil
| | - Francisco Izaias da Silva Aires
- Instituto de Engenharias e Desenvolvimento Sustentável, Universidade da Integração Internacional da Lusofonia Afro-Brasileira Campus das Auroras Redenção CEP 62790970 CE Brazil
| | - Erico Carlos de Castro
- Departamento de Química Orgânica e Inorgânica, Centro de Ciências, Universidade Federal do Ceará Campus Pici Fortaleza CEP 60455760 CE Brazil
| | - Kaiany Moreira Dos Santos
- Instituto de Engenharias e Desenvolvimento Sustentável, Universidade da Integração Internacional da Lusofonia Afro-Brasileira Campus das Auroras Redenção CEP 62790970 CE Brazil
| | - José Cleiton Sousa Dos Santos
- Departamento de Química Orgânica e Inorgânica, Centro de Ciências, Universidade Federal do Ceará Campus Pici Fortaleza CEP 60455760 CE Brazil
- Instituto de Engenharias e Desenvolvimento Sustentável, Universidade da Integração Internacional da Lusofonia Afro-Brasileira Campus das Auroras Redenção CEP 62790970 CE Brazil
- Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará Campus do Pici, Bloco 940 Fortaleza CEP 60455760 CE Brazil
| |
Collapse
|
8
|
Vergani-Junior CA, Moro RDP, Pinto S, De-Souza EA, Camara H, Braga DL, Tonon-da-Silva G, Knittel TL, Ruiz GP, Ludwig RG, Massirer KB, Mair WB, Mori MA. An Intricate Network Involving the Argonaute ALG-1 Modulates Organismal Resistance to Oxidative Stress. Nat Commun 2024; 15:3070. [PMID: 38594249 PMCID: PMC11003958 DOI: 10.1038/s41467-024-47306-4] [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: 08/01/2023] [Accepted: 03/24/2024] [Indexed: 04/11/2024] Open
Abstract
Cellular response to redox imbalance is crucial for organismal health. microRNAs are implicated in stress responses. ALG-1, the C. elegans ortholog of human AGO2, plays an essential role in microRNA processing and function. Here we investigated the mechanisms governing ALG-1 expression in C. elegans and the players controlling lifespan and stress resistance downstream of ALG-1. We show that upregulation of ALG-1 is a shared feature in conditions linked to increased longevity (e.g., germline-deficient glp-1 mutants). ALG-1 knockdown reduces lifespan and oxidative stress resistance, while overexpression enhances survival against pro-oxidant agents but not heat or reductive stress. R02D3.7 represses alg-1 expression, impacting oxidative stress resistance at least in part via ALG-1. microRNAs upregulated in glp-1 mutants (miR-87-3p, miR-230-3p, and miR-235-3p) can target genes in the protein disulfide isomerase pathway and protect against oxidative stress. This study unveils a tightly regulated network involving transcription factors and microRNAs which controls organisms' ability to withstand oxidative stress.
Collapse
Affiliation(s)
- Carlos A Vergani-Junior
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Raíssa De P Moro
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Silas Pinto
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Evandro A De-Souza
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Henrique Camara
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Section on Integrative Physiology & Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - Deisi L Braga
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Guilherme Tonon-da-Silva
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Thiago L Knittel
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Gabriel P Ruiz
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Raissa G Ludwig
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Katlin B Massirer
- Center for Molecular Biology and Genetic Engineering (CBMEG), Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
- Center of Medicinal Chemistry (CQMED), Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - William B Mair
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil.
- Program in Genetics and Molecular Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil.
- Obesity and Comorbidities Research Center (OCRC), Universidade Estadual de Campinas, Campinas, São Paulo, Brazil.
- Experimental Medicine Research Cluster (EMRC), Universidade Estadual de Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
9
|
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
|
10
|
Wang Y, Yuan T, Wang H, Meng Q, Li H, Feng C, Li Z, Sun S. Inhibition of Protein Disulfide Isomerase Attenuates Osteoclast Differentiation and Function via the Readjustment of Cellular Redox State in Postmenopausal Osteoporosis. Inflammation 2024; 47:626-648. [PMID: 38055120 DOI: 10.1007/s10753-023-01933-z] [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: 08/29/2023] [Revised: 10/16/2023] [Accepted: 11/13/2023] [Indexed: 12/07/2023]
Abstract
Due to the accumulation of reactive oxygen species (ROS) and heightened activity of osteoclasts, postmenopausal osteoporosis could cause severe pathological bone destruction. Protein disulfide isomerase (PDI), an endoplasmic prototypic thiol isomerase, plays a central role in affecting cellular redox state. To test whether suppression of PDI could inhibit osteoclastogenesis through cellular redox regulation, bioinformatics network analysis was performed on the causative genes, followed by biological validation on the osteoclastogenesis in vitro and ovariectomy (OVX) mice model in vivo. The analysis identified PDI as one of gene targets for postmenopausal osteoporosis, which was positively expressed during osteoclastogenesis. Therefore, PDI expression inhibitor and chaperone activity inhibitor were used to verify the effects of PDI inhibitors on osteoclastogenesis. Results demonstrated that PDI inhibitors could reduce osteoclast number and inhibit resorption function via suppression on osteoclast marker genes. The mechanisms behind the scenes were the PDI inhibitors-caused intracellular ROS reduction via enhancement of the antioxidant system. Micro-CT and histological results indicated PDI inhibitors could effectively alleviate or even prevent bone loss in OVX mice. In conclusion, our findings unveiled the suppressive effects of PDI inhibitors on osteoclastogenesis by reducing intracellular ROS, providing new therapeutic options for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Yi Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Tao Yuan
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Haojue Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Qi Meng
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Haoyang Li
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Changgong Feng
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Ziqing Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| |
Collapse
|
11
|
Moretti AIS, Baksheeva VE, Roman AY, De Bessa TC, Devred F, Kovacic H, Tsvetkov PO. Exploring the Influence of Zinc Ions on the Conformational Stability and Activity of Protein Disulfide Isomerase. Int J Mol Sci 2024; 25:2095. [PMID: 38396772 PMCID: PMC10889200 DOI: 10.3390/ijms25042095] [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: 11/28/2023] [Revised: 01/22/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
The interplay between metal ion binding and the activity of thiol proteins, particularly within the protein disulfide isomerase family, remains an area of active investigation due to the critical role that these proteins play in many vital processes. This research investigates the interaction between recombinant human PDIA1 and zinc ions, focusing on the subsequent implications for PDIA1's conformational stability and enzymatic activity. Employing isothermal titration calorimetry and differential scanning calorimetry, we systematically compared the zinc binding capabilities of both oxidized and reduced forms of PDIA1 and assessed the structural consequences of this interaction. Our results demonstrate that PDIA1 can bind zinc both in reduced and oxidized states, but with significantly different stoichiometry and more pronounced conformational effects in the reduced form of PDIA1. Furthermore, zinc binding was observed to inhibit the catalytic activity of reduced-PDIA1, likely due to induced alterations in its conformation. These findings unveil a potential regulatory mechanism in PDIA1, wherein metal ion binding under reductive conditions modulates its activity. Our study highlights the potential role of zinc in regulating the catalytic function of PDIA1 through conformational modulation, suggesting a nuanced interplay between metal binding and protein stability in the broader context of cellular redox regulation.
Collapse
Affiliation(s)
- Ana Iochabel Soares Moretti
- Vascular Biology Laboratory (LIM64), School of Medicine, Heart Institute (InCor), Cardiopneumology Department, University of São Paulo, Campus Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Viktoria E. Baksheeva
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Fac Sciences Médicales et Paramédicales, 13005 Marseille, France (F.D.); (H.K.)
| | - Andrei Yu. Roman
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Fac Sciences Médicales et Paramédicales, 13005 Marseille, France (F.D.); (H.K.)
| | - Tiphany Coralie De Bessa
- Vascular Biology Laboratory (LIM64), School of Medicine, Heart Institute (InCor), Cardiopneumology Department, University of São Paulo, Campus Sao Paulo, Sao Paulo 05403-000, Brazil
| | - François Devred
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Fac Sciences Médicales et Paramédicales, 13005 Marseille, France (F.D.); (H.K.)
| | - Hervé Kovacic
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Fac Sciences Médicales et Paramédicales, 13005 Marseille, France (F.D.); (H.K.)
| | - Philipp O. Tsvetkov
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Fac Sciences Médicales et Paramédicales, 13005 Marseille, France (F.D.); (H.K.)
| |
Collapse
|
12
|
Trevisan R, Mello DF. Redox control of antioxidants, metabolism, immunity, and development at the core of stress adaptation of the oyster Crassostrea gigas to the dynamic intertidal environment. Free Radic Biol Med 2024; 210:85-106. [PMID: 37952585 DOI: 10.1016/j.freeradbiomed.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
This review uses the marine bivalve Crassostrea gigas to highlight redox reactions and control systems in species living in dynamic intertidal environments. Intertidal species face daily and seasonal environmental variability, including temperature, oxygen, salinity, and nutritional changes. Increasing anthropogenic pressure can bring pollutants and pathogens as additional stressors. Surprisingly, C. gigas demonstrates impressive adaptability to most of these challenges. We explore how ROS production, antioxidant protection, redox signaling, and metabolic adjustments can shed light on how redox biology supports oyster survival in harsh conditions. The review provides (i) a brief summary of shared redox sensing processes in metazoan; (ii) an overview of unique characteristics of the C. gigas intertidal habitat and the suitability of this species as a model organism; (iii) insights into the redox biology of C. gigas, including ROS sources, signaling pathways, ROS-scavenging systems, and thiol-containing proteins; and examples of (iv) hot topics that are underdeveloped in bivalve research linking redox biology with immunometabolism, physioxia, and development. Given its plasticity to environmental changes, C. gigas is a valuable model for studying the role of redox biology in the adaptation to harsh habitats, potentially providing novel insights for basic and applied studies in marine and comparative biochemistry and physiology.
Collapse
Affiliation(s)
- Rafael Trevisan
- Univ Brest, Ifremer, CNRS, IRD, UMR 6539, LEMAR, Plouzané, 29280, France
| | - Danielle F Mello
- Univ Brest, Ifremer, CNRS, IRD, UMR 6539, LEMAR, Plouzané, 29280, France.
| |
Collapse
|
13
|
Porto FG, Tanaka LY, de Bessa TC, Oliveira PVS, Souza JMFD, Kajihara D, Fernandes CG, Santos PN, Laurindo FRM. Evidence for a protective role of Protein Disulfide Isomerase-A1 against aortic dissection. Atherosclerosis 2023; 382:117283. [PMID: 37774430 DOI: 10.1016/j.atherosclerosis.2023.117283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/25/2023] [Accepted: 09/06/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND AND AIMS Redox signaling is involved in the pathophysiology of aortic aneurysm/dissection. Protein Disulfide Isomerases and its prototype PDIA1 are thiol redox chaperones mainly from endoplasmic reticulum (ER), while PDIA1 cell surface pool redox-regulates thrombosis, cytoskeleton remodeling and integrin activation, which are mechanisms involved in aortic disease. Here we investigate the roles of PDIA1 in aortic dissection. METHODS Initially, we assessed the outcome of aortic aneurysm/dissection in transgenic PDIA1-overexpressing FVB mice using a model of 28-day exposure to lysyl oxidase inhibitor BAPN plus angiotensin-II infusion. In a second protocol, we assessed the effects of PDIA1 inhibitor isoquercetin (IQ) against aortic dissection in C57BL/6 mice exposed to BAPN for 28 days. RESULTS Transgenic PDIA1 overexpression associated with ca. 50% (p = 0.022) decrease (vs.wild-type) in mortality due to abdominal aortic rupture and protected against elastic fiber breaks in thoracic aorta. Conversely, exposure of mice to IQ increased thoracic aorta dissection-related mortality rates, from ca. 18%-50% within 28-days (p = 0.019); elastic fiber disruption and collagen deposition were also enhanced. The structurally-related compound diosmetin, which does not inhibit PDI, had negligible effects. In parallel, stretch-tension curves indicated that IQ amplified a ductile-type of biomechanical failure vs. control or BAPN-exposed mice aortas. IQ-induced effects seemed unassociated with nonspecific antioxidant effects or ER stress. In both models, echocardiographic analysis of surviving mice suggested that aortic rupture was dissociated from progressive dilatation. CONCLUSIONS Our data indicate a protective role of PDIA1 against aortic dissection/rupture and potentially uncovers a novel integrative mechanism coupling redox and biomechanical homeostasis in vascular remodeling.
Collapse
Affiliation(s)
- Fernando Garcez Porto
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Leonardo Yuji Tanaka
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, 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 (LVascBio), 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
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Júlia Martins Felipe de Souza
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Daniela Kajihara
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Carolina Gonçalves Fernandes
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Patricia Nolasco Santos
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Francisco Rafael Martins Laurindo
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
14
|
Peng XX, Zhang H, Zhang R, Li ZH, Yang ZS, Zhang J, Gao S, Zhang JL. Gallium Triggers Ferroptosis through a Synergistic Mechanism. Angew Chem Int Ed Engl 2023; 62:e202307838. [PMID: 37452698 DOI: 10.1002/anie.202307838] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/18/2023]
Abstract
The gallium ion (Ga3+ ) has long been believed to disrupt ferric homeostasis in the body by competing with iron cofactors in metalloproteins, ultimately leading to cell death. This study revealed that through an indirect pathway, gallium can trigger ferroptosis, a type of non-apoptotic cell death regulated by iron. This is exemplified by the gallium complex of the salen ligand (Ga-1); we found that Ga-1 acts as an effective anion transporter that can affect the pH gradient and change membrane permeability, leading to mitochondrial dysfunction and the release of ferrous iron from the electron transfer chain (ETC). In addition, Ga-1 also targeted protein disulfide isomerases (PDIs) located in the endoplasmic reticulum (ER) membrane, preventing the repair of the antioxidant glutathione (GSH) system and thus enforcing ferroptosis. Finally, a combination treatment of Ga-1 and dietary polyunsaturated fatty acids (PUFAs), which enhances lipid peroxidation during ferroptosis, showed a synergistic therapeutic effect both in vitro and in vivo. This study provided us with a strategy to synergistically induce Ferroptosis in tumor cells, thereby enhancing the anti-neoplastic effect.
Collapse
Affiliation(s)
- Xin-Xin Peng
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Hang Zhang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Ruijing Zhang
- Spin-X Institute, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
| | - Ze-Hao Li
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zi-Shu Yang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Jing Zhang
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Song Gao
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
- Spin-X Institute, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, 515031, P. R. China
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Jun-Long Zhang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, 515031, P. R. China
| |
Collapse
|
15
|
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
|
16
|
Nagarkoti S, Kim YM, Ash D, Das A, Vitriol E, Read TA, Youn SW, Sudhahar V, McMenamin M, Hou Y, Boatwright H, Caldwell R, Essex DW, Cho J, Fukai T, Ushio-Fukai M. Protein disulfide isomerase A1 as a novel redox sensor in VEGFR2 signaling and angiogenesis. Angiogenesis 2023; 26:77-96. [PMID: 35984546 PMCID: PMC9918675 DOI: 10.1007/s10456-022-09852-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 07/26/2022] [Indexed: 02/04/2023]
Abstract
VEGFR2 signaling in endothelial cells (ECs) is regulated by reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) and mitochondria, which plays an important role in postnatal angiogenesis. However, it remains unclear how highly diffusible ROS signal enhances VEGFR2 signaling and reparative angiogenesis. Protein disulfide isomerase A1 (PDIA1) functions as an oxidoreductase depending on the redox environment. We hypothesized that PDIA1 functions as a redox sensor to enhance angiogenesis. Here we showed that PDIA1 co-immunoprecipitated with VEGFR2 or colocalized with either VEGFR2 or an early endosome marker Rab5 at the perinuclear region upon stimulation of human ECs with VEGF. PDIA1 silencing significantly reduced VEGF-induced EC migration, proliferation and spheroid sprouting via inhibiting VEGFR2 signaling. Mechanistically, VEGF stimulation rapidly increased Cys-OH formation of PDIA1 via the NOX4-mitochondrial ROS axis. Overexpression of "redox-dead" mutant PDIA1 with replacement of the active four Cys residues with Ser significantly inhibited VEGF-induced PDIA1-CysOH formation and angiogenic responses via reducing VEGFR2 phosphorylation. Pdia1+/- mice showed impaired angiogenesis in developmental retina and Matrigel plug models as well as ex vivo aortic ring sprouting model. Study using hindlimb ischemia model revealed that PDIA1 expression was markedly increased in angiogenic ECs of ischemic muscles, and that ischemia-induced limb perfusion recovery and neovascularization were impaired in EC-specific Pdia1 conditional knockout mice. These results suggest that PDIA1 can sense VEGF-induced H2O2 signal via CysOH formation to promote VEGFR2 signaling and angiogenesis in ECs, thereby enhancing postnatal angiogenesis. The oxidized PDIA1 is a potential therapeutic target for treatment of ischemic vascular diseases.
Collapse
Affiliation(s)
- Sheela Nagarkoti
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
| | - Young-Mee Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Department of Medicine (Cardiology), University of Illinois at Chicago, Chicago, IL, USA
| | - Dipankar Ash
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
| | - Archita Das
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
| | - Eric Vitriol
- Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Tracy-Ann Read
- Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Seock-Won Youn
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Varadarajan Sudhahar
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA
| | - Malgorzata McMenamin
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA
| | - Yali Hou
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA
| | - Harriet Boatwright
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
| | - Ruth Caldwell
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Vision Discovery Institute, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA
| | - David W Essex
- Department of Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Departments of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA.
- Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
17
|
Dousti M, Hosseinpour M, D Ghasemi N, Mirfakhraee H, Rajabi SK, Rashidi S, Hatam G. The potential role of protein disulfide isomerases (PDIs) during parasitic infections: a focus on Leishmania spp. Pathog Dis 2023; 81:ftad032. [PMID: 38061803 DOI: 10.1093/femspd/ftad032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/24/2023] [Accepted: 12/05/2023] [Indexed: 12/22/2023] Open
Abstract
Leishmaniasis is a group of vector-borne diseases caused by intracellular protozoan parasites belonging to the genus Leishmania. Leishmania parasites can employ different and numerous sophisticated strategies, including modulating host proteins, cell signaling, and cell responses by parasite proteins, to change the infected host conditions to favor the parasite persistence and induce pathogenesis. In this sense, protein disulfide isomerases (PDIs) have been described as crucial proteins that can be modulated during leishmaniasis and affect the pathogenesis process. The effect of modulated PDIs can be investigated in both aspects, parasite PDIs and infected host cell PDIs, during infection. The information concerning PDIs is not sufficient in parasitology; however, this study aimed to provide data regarding the biological functions of such crucial proteins in parasites with a focus on Leishmania spp. and their relevant effects on the pathogenesis process. Although there are no clinical trial vaccines and therapeutic approaches, highlighting this information might be fruitful for the development of novel strategies based on PDIs for the management of parasitic diseases, especially leishmaniasis.
Collapse
Affiliation(s)
- Majid Dousti
- Firoozabadi Clinical Research Development Unit (FACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Hosseinpour
- Student Research Committee, School of Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Nadia D Ghasemi
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hosna Mirfakhraee
- Firoozabadi Clinical Research Development Unit (FACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Shahin K Rajabi
- Firoozabadi Clinical Research Development Unit (FACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Rashidi
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran
- Department of Medical Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran
| | - Gholamreza Hatam
- Basic Sciences Infectious Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
18
|
Kong X, Yao H, Ren J, Dai W, Lin Z, Li C, Dong Y. PDIA6 involves the thermal stress response of razor clam, Sinonovacula constricta. FISH & SHELLFISH IMMUNOLOGY 2022; 131:766-774. [PMID: 36349651 DOI: 10.1016/j.fsi.2022.10.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/11/2022] [Accepted: 10/24/2022] [Indexed: 06/16/2023]
Abstract
Protein disulfide isomerases A6 (PDIA6), an oxidoreductase and isomerase, catalyzes the oxidation reduction and isomerization of disulfide bonds, and serves as molecular chaperone to prevent the buildup of misfolded proteins under various environmental insults. However, the role of PDIA6 in mollusks remains largely obscure, although its multifunctional protein has been reported in other species under adverse conditions. To fill this gap, we identified PDIA6 from the razor clam Sinonovacula constricta (ScPDIA6) and investigated its expression patterns in response to thermal stress. Tissue distribution showed that the mRNA transcript of ScPDIA6 was ubiquitously expressed in nine tested tissues. Temporal expression profiles by qPCR revealed that ScPDIA6 in the gill and mantle was significantly increased by hyper-thermic treatment. Further, Western blot and immunofluorescence indicated that ScPDIA6 was significantly upregulated by thermal treatment at the protein level. Additionally, the survival test demonstrated that the viability of E. coli cells expressing recombinant ScPDIA6 protein increased at 42 °C compared with empty vector. Overall, these findings suggested that ScPDIA6 may play a pivotal role in counteracting thermal stress. This study will provide valuable reference data resource for understanding the potential role of PDIA6 in mollusks.
Collapse
Affiliation(s)
- Xianghui Kong
- College of Marine Sciences, Ningbo University, Ningbo, 315010, PR China; Zhejiang Key Laboratory of Aquatic Germplasm Resource, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, PR China
| | - Hanhan Yao
- Zhejiang Key Laboratory of Aquatic Germplasm Resource, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, PR China
| | - Jianfeng Ren
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, PR China
| | - Wenfang Dai
- Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ninghai, 315604, PR China
| | - Zhihua Lin
- Zhejiang Key Laboratory of Aquatic Germplasm Resource, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, PR China; Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ninghai, 315604, PR China
| | - Chenghua Li
- College of Marine Sciences, Ningbo University, Ningbo, 315010, PR China.
| | - Yinghui Dong
- Zhejiang Key Laboratory of Aquatic Germplasm Resource, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, PR China; Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ninghai, 315604, PR China.
| |
Collapse
|
19
|
Tong S, Yin C, Ge Y, Ren Z, Tao J, Liu Y. Albumin (ALB) and protein disulfide isomerase family A member 4 (PDIA4) are novel markers to predict sperm freezability of Erhualian boar. Cryobiology 2022; 109:37-43. [PMID: 36181861 DOI: 10.1016/j.cryobiol.2022.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 01/16/2023]
Abstract
The frozen semen of Erhualian pig can promote the continuous improvement of commercial pigs, but currently, frozen semen fails to satisfy the practical application requirement. Oxidative damage is one of the crucial factors affecting the quality of frozen semen; besides, there are individual differences in boar sperm freezability. Based on the previous analysis of the proteomic differences of Erhualian boar sperm with different freezability, two differentially abundant proteins (DAPs) in boar sperm, albumin (ALB) and protein disulfide isomerase family A member 4 (PDIA4), were selected as the research objects in the current study. It is assumed that redox-related proteins ALB and PDIA4 can be used as markers to predict Erhualian boar sperm freezability. We cryopreserved the semen of 14 Erhualian boars. According to the difference of frozen semen quality, boars with good and poor freezability ejaculates (GFE and PFE, n = 3) were selected respectively. The relative contents of ALB and PDIA4 in GFE and PFE were analyzed by Western blot, and the localization patterns of ALB and PDIA4 in pre-frozen and frozen-thawed sperm were detected by immunofluorescence. The results showed that the abundances of ALB and PDIA4 in GFE were significantly higher than PFE, and there was a significant correlation between the relative contents of ALB and PDIA4 and frozen-thawed sperm quality parameters. Additionally, the freezing process had no effect on the localization patterns of ALB and PDIA4 in spermatozoa. In conclusion, these results suggest that ALB and PDIA4 are related to boar sperm cryotolerance and may be used as novel freezability markers.
Collapse
Affiliation(s)
- Shifeng Tong
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Chang Yin
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Yuzhu Ge
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Zhibin Ren
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Jingli Tao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Yang Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, 210095, PR China.
| |
Collapse
|
20
|
Hung CT, Su TH, Chen YT, Wu YF, Chen YT, Lin SJ, Lin SL, Yang KC. Targeting ER protein TXNDC5 in hepatic stellate cell mitigates liver fibrosis by repressing non-canonical TGFβ signalling. Gut 2022; 71:1876-1891. [PMID: 34933915 DOI: 10.1136/gutjnl-2021-325065] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 12/05/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND OBJECTIVES Liver fibrosis (LF) occurs following chronic liver injuries. Currently, there is no effective therapy for LF. Recently, we identified thioredoxin domain containing 5 (TXNDC5), an ER protein disulfide isomerase (PDI), as a critical mediator of cardiac and lung fibrosis. We aimed to determine if TXNDC5 also contributes to LF and its potential as a therapeutic target for LF. DESIGN Histological and transcriptome analyses on human cirrhotic livers were performed. Col1a1-GFPTg , Alb-Cre;Rosa26-tdTomato and Tie2-Cre/ERT2;Rosa26-tdTomato mice were used to determine the cell type(s) where TXNDC5 was induced following liver injury. In vitro investigations were conducted in human hepatic stellate cells (HSCs). Col1a2-Cre/ERT2;Txndc5fl/fl (Txndc5cKO ) and Alb-Cre;Txndc5fl/fl (Txndc5Hep-cKO ) mice were generated to delete TXNDC5 in HSCs and hepatocytes, respectively. Carbon tetrachloride treatment and bile duct ligation surgery were employed to induce liver injury/fibrosis in mice. The extent of LF was quantified using histological, imaging and biochemical analyses. RESULTS TXNDC5 was upregulated markedly in human and mouse fibrotic livers, particularly in activated HSC at the fibrotic foci. TXNDC5 was induced by transforming growth factor β1 (TGFβ1) in HSCs and it was both required and sufficient for the activation, proliferation, survival and extracellular matrix production of HSC. Mechanistically, TGFβ1 induces TXNDC5 expression through increased ER stress and ATF6-mediated transcriptional regulation. In addition, TXNDC5 promotes LF by redox-dependent JNK and signal transducer and activator of transcription 3 activation in HSCs through its PDI activity, activating HSCs and making them resistant to apoptosis. HSC-specific deletion of Txndc5 reverted established LF in mice. CONCLUSIONS ER protein TXNDC5 promotes LF through redox-dependent HSC activation, proliferation and excessive extracellular matrix production. Targeting TXNDC5, therefore, could be a potential novel therapeutic strategy to ameliorate LF.
Collapse
Affiliation(s)
- Chen-Ting Hung
- Graduate Institute and Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tung-Hung Su
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Ting Chen
- Graduate Institute and Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yueh-Feng Wu
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - You-Tzung Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sung-Jan Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.,Department of Dermatology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.,Research Center for Developmental Biology & Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Center for Frontier Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shuei-Liong Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Research Center for Developmental Biology & Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute and Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, Taipei, Taiwan
| | - Kai-Chien Yang
- Graduate Institute and Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan .,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Research Center for Developmental Biology & Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
21
|
Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers (Basel) 2022; 14:cancers14174082. [PMID: 36077618 PMCID: PMC9454959 DOI: 10.3390/cancers14174082] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.
Collapse
Affiliation(s)
- Danai Dima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongxu Jiang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Divya Jyoti Singh
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Haikoo S. Shah
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
22
|
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
|
23
|
Palazzo FC, Sitia R, Tempio T. Selective Secretion of KDEL-Bearing Proteins: Mechanisms and Functions. Front Cell Dev Biol 2022; 10:967875. [PMID: 35912099 PMCID: PMC9326092 DOI: 10.3389/fcell.2022.967875] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
In multicellular organisms, cells must continuously exchange messages with the right meaning, intensity, and duration. Most of these messages are delivered through cognate interactions between membrane and secretory proteins. Their conformational maturation is assisted by a vast array of chaperones and enzymes, ensuring the fidelity of intercellular communication. These folding assistants reside in the early secretory compartment (ESC), a functional unit that encompasses endoplasmic reticulum (ER), intermediate compartment and cis-Golgi. Most soluble ESC residents have C-terminal KDEL-like motifs that prevent their transport beyond the Golgi. However, some accumulate in the ER, while others in downstream stations, implying different recycling rates. Moreover, it is now clear that cells can actively secrete certain ESC residents but not others. This essay discusses the physiology of their differential intracellular distribution, and the mechanisms that may ensure selectivity of release.
Collapse
|
24
|
Karcini A, Lazar IM. The SKBR3 cell-membrane proteome reveals telltales of aberrant cancer cell proliferation and targets for precision medicine applications. Sci Rep 2022; 12:10847. [PMID: 35760832 PMCID: PMC9237123 DOI: 10.1038/s41598-022-14418-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/07/2022] [Indexed: 12/14/2022] Open
Abstract
The plasma membrane proteome resides at the interface between the extra- and intra-cellular environment and through its various roles in signal transduction, immune recognition, nutrient transport, and cell-cell/cell-matrix interactions plays an absolutely critical role in determining the fate of a cell. Our work was aimed at exploring the cell-membrane proteome of a HER2+ breast-cancer cell line (SKBR3) to identify triggers responsible for uncontrolled cell proliferation and intrinsic resources that enable detection and therapeutic interventions. To mimic environmental conditions that enable cancer cells to evolve adaptation/survival traits, cell culture was performed under serum-rich and serum-deprived conditions. Proteomic analysis enabled the identification of ~ 2000 cell-membrane proteins. Classification into proteins with receptor/enzymatic activity, CD antigens, transporters, and cell adhesion/junction proteins uncovered overlapping roles in processes that drive cell growth, apoptosis, differentiation, immune response, adhesion and migration, as well as alternate pathways for proliferation. The large number of tumor markers (> 50) and putative drug targets (> 100) exposed a vast potential for yet unexplored detection and targeting opportunities, whereas the presence of 15 antigen immunological markers enabled an assessment of epithelial, mesenchymal or stemness characteristics. Serum-starved cells displayed altered processes related to mitochondrial OXPHOS/ATP synthesis, protein folding and localization, while serum-treated cells exhibited attributes that support tissue invasion and metastasis. Altogether, our findings advance the understanding of the biological triggers that sustain aberrant cancer cell proliferation, survival and development of resistance to therapeutic drugs, and reveal vast innate opportunities for guiding immunological profiling and precision medicine applications aimed at target selection or drug discovery.
Collapse
Affiliation(s)
- Arba Karcini
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Iulia M Lazar
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA.
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA.
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA.
| |
Collapse
|
25
|
The proteomics of the freshwater pearl powder: Insights from biomineralization to biomedical application. J Proteomics 2022; 265:104665. [PMID: 35753678 DOI: 10.1016/j.jprot.2022.104665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/30/2022] [Accepted: 06/15/2022] [Indexed: 11/24/2022]
Abstract
The freshwater pearl is one kind of valuable organic jewelry and traditional Chinese medicine (TCM). However, the molecular basis of matrix protein in pearl biomineralization and biomedical applications are largely unknown to date. In this study, the matrix proteins of water-soluble matrix, acid-soluble matrix and acid-insoluble matrix from the freshwater seedless pearl powder were detected using liquid chromatography-tandem mass spectrometry (LC-MS/MS) respectively, and identified against the transcriptomic database of the pearl sac. The results showed that a total of 190 proteins were identified in pearl proteomics, which was divided into eight categories by their potential biomineralization functions. The composition of pearl matrix proteins and the high frequency conserved domains like carbonic anhydrase, von Willebrand factor type A, tyrosinase and chitin binding 2 in protein sequences, implying that the "chitin-silk fibroin gel proteins-acidic macromolecules" model was suitable for description the pearl biomineralization process. Meanwhile, ninety-one of pearl matrix proteins could be classified into seven categories by their potential medical functions including wound healing, osteogenic property, antioxidant activity, neuro-regulation effects, skin lightening effect, anti-inflammatory and anti-apoptotic effects and other immunomodulatory property. In general, these results provided valuable new insights into not only the diversity of pearl matrix protein for mollusc biomineralization, but the molecular basis of pearl matrix proteins responsible for their diverse biological properties in TCM application. SIGNIFICANCE: The significance of this study included the following points.
Collapse
|
26
|
Domesticated LTR-Retrotransposon gag-Related Gene (Gagr) as a Member of the Stress Response Network in Drosophila. Life (Basel) 2022; 12:life12030364. [PMID: 35330115 PMCID: PMC8956099 DOI: 10.3390/life12030364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/27/2022] [Accepted: 02/27/2022] [Indexed: 11/24/2022] Open
Abstract
The most important sources of new components of genomes are transposable elements, which can occupy more than half of the nucleotide sequence of the genome in higher eukaryotes. Among the mobile components of a genome, a special place is occupied by retroelements, which are similar to retroviruses in terms of their mechanisms of integration into a host genome. The process of positive selection of certain sequences of transposable elements and retroviruses in a host genome is commonly called molecular domestication. There are many examples of evolutionary adaptations of gag (retroviral capsid) sequences as new regulatory sequences of different genes in mammals, where domesticated gag genes take part in placenta functioning and embryogenesis, regulation of apoptosis, hematopoiesis, and metabolism. The only gag-related gene has been found in the Drosophila genome—Gagr. According to the large-scale transcriptomic and proteomic analysis data, the Gagr gene in D. melanogaster is a component of the protein complex involved in the stress response. In this work, we consider the evolutionary processes that led to the formation of a new function of the domesticated gag gene and its adaptation to participation in the stress response. We discuss the possible functional role of the Gagr as part of the complex with its partners in Drosophila, and the pathway of evolution of proteins of the complex in eukaryotes to determine the benefit of the domesticated retroelement gag gene.
Collapse
|
27
|
Carvalho LAC, Queijo RG, Baccaro ALB, Siena ÁDD, Silva WA, Rodrigues T, Maria-Engler SS. Redox-Related Proteins in Melanoma Progression. Antioxidants (Basel) 2022; 11:438. [PMID: 35326089 PMCID: PMC8944639 DOI: 10.3390/antiox11030438] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/05/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Melanoma is the most aggressive type of skin cancer. Despite the available therapies, the minimum residual disease is still refractory. Reactive oxygen and nitrogen species (ROS and RNS) play a dual role in melanoma, where redox imbalance is involved from initiation to metastasis and resistance. Redox proteins modulate the disease by controlling ROS/RNS levels in immune response, proliferation, invasion, and relapse. Chemotherapeutics such as BRAF and MEK inhibitors promote oxidative stress, but high ROS/RNS amounts with a robust antioxidant system allow cells to be adaptive and cooperate to non-toxic levels. These proteins could act as biomarkers and possible targets. By understanding the complex mechanisms involved in adaptation and searching for new targets to make cells more susceptible to treatment, the disease might be overcome. Therefore, exploring the role of redox-sensitive proteins and the modulation of redox homeostasis may provide clues to new therapies. This study analyzes information obtained from a public cohort of melanoma patients about the expression of redox-generating and detoxifying proteins in melanoma during the disease stages, genetic alterations, and overall patient survival status. According to our analysis, 66% of the isoforms presented differential expression on melanoma progression: NOS2, SOD1, NOX4, PRX3, PXDN and GPX1 are increased during melanoma progression, while CAT, GPX3, TXNIP, and PRX2 are decreased. Besides, the stage of the disease could influence the result as well. The levels of PRX1, PRX5 and PRX6 can be increased or decreased depending on the stage. We showed that all analyzed isoforms presented some genetic alteration on the gene, most of them (78%) for increased mRNA expression. Interestingly, 34% of all melanoma patients showed genetic alterations on TRX1, most for decreased mRNA expression. Additionally, 15% of the isoforms showed a significant reduction in overall patient survival status for an altered group (PRX3, PRX5, TR2, and GR) and the unaltered group (NOX4). Although no such specific antioxidant therapy is approved for melanoma yet, inhibitors or mimetics of these redox-sensitive proteins have achieved very promising results. We foresee that forthcoming investigations on the modulation of these proteins will bring significant advances for cancer therapy.
Collapse
Affiliation(s)
- Larissa A. C. Carvalho
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo 05508-00, SP, Brazil; (L.A.C.C.); (R.G.Q.)
| | - Rodrigo G. Queijo
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo 05508-00, SP, Brazil; (L.A.C.C.); (R.G.Q.)
| | - Alexandre L. B. Baccaro
- Centro de Pós-Graduação e Pesquisa Oswaldo Cruz, Faculdade Oswaldo Cruz, Rua Brigadeiro Galvão, 535, Sao Paulo 01151-000, SP, Brazil;
| | - Ádamo D. D. Siena
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900, Ribeirao Preto 14049-900, SP, Brazil; (Á.D.D.S.); (W.A.S.J.)
| | - Wilson A. Silva
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900, Ribeirao Preto 14049-900, SP, Brazil; (Á.D.D.S.); (W.A.S.J.)
| | - Tiago Rodrigues
- Center for Natural and Human Sciences, Federal University of ABC, Avenida dos Estados, 5001, Santo Andre 09210-580, SP, Brazil;
| | - Silvya Stuchi Maria-Engler
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo 05508-00, SP, Brazil; (L.A.C.C.); (R.G.Q.)
| |
Collapse
|
28
|
Arsiccio A, Metcalfe C, Pisano R, Raut S, Coxon C. A proximity-based in silico approach to identify redox-labile disulfide bonds: The example of FVIII. PLoS One 2022; 17:e0262409. [PMID: 35130281 PMCID: PMC8820644 DOI: 10.1371/journal.pone.0262409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/27/2021] [Indexed: 01/04/2023] Open
Abstract
Allosteric disulfide bonds permit highly responsive, transient 'switch-like' properties that are ideal for processes like coagulation and inflammation that require rapid and localised responses to damage or injury. Haemophilia A (HA) is a rare bleeding disorder managed with exogenous coagulation factor(F) VIII products. FVIII has eight disulfide bonds and is known to be redox labile, but it is not known how reduction/oxidation affects the structure-function relationship, or its immunogenicity-a serious complication for 30% severe HA patients. Understanding how redox-mediated changes influence FVIII can inform molecular engineering strategies aimed at improving activity and stability, and reducing immunogenicity. FVIII is a challenging molecule to work with owing to its poor expression and instability so, in a proof-of-concept study, we used molecular dynamics (MD) to identify which disulfide bonds were most likely to be reduced and how this would affect structure/function; results were then experimentally verified. MD identified Cys1899-Cys1903 disulfide as the most likely to undergo reduction based on energy and proximity criteria. Further MD suggested this reduction led to a more open conformation. Here we present our findings and highlight the value of MD approaches.
Collapse
Affiliation(s)
- Andrea Arsiccio
- Department of Applied Science and Technology, Politecnico di Torino, Torino, Italy
| | - Clive Metcalfe
- National Institute for Biological Standards and Control, Hertfordshire, United Kingdom
| | - Roberto Pisano
- Department of Applied Science and Technology, Politecnico di Torino, Torino, Italy
| | - Sanj Raut
- National Institute for Biological Standards and Control, Hertfordshire, United Kingdom
| | - Carmen Coxon
- National Institute for Biological Standards and Control, Hertfordshire, United Kingdom
| |
Collapse
|
29
|
Wen MH, Xie X, Huang PS, Yang K, Chen TY. Crossroads between membrane trafficking machinery and copper homeostasis in the nerve system. Open Biol 2021; 11:210128. [PMID: 34847776 PMCID: PMC8633785 DOI: 10.1098/rsob.210128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Imbalanced copper homeostasis and perturbation of membrane trafficking are two common symptoms that have been associated with the pathogenesis of neurodegenerative and neurodevelopmental diseases. Accumulating evidence from biophysical, cellular and in vivo studies suggest that membrane trafficking orchestrates both copper homeostasis and neural functions-however, a systematic review of how copper homeostasis and membrane trafficking interplays in neurons remains lacking. Here, we summarize current knowledge of the general trafficking itineraries for copper transporters and highlight several critical membrane trafficking regulators in maintaining copper homeostasis. We discuss how membrane trafficking regulators may alter copper transporter distribution in different membrane compartments to regulate intracellular copper homeostasis. Using Parkinson's disease and MEDNIK as examples, we further elaborate how misregulated trafficking regulators may interplay parallelly or synergistically with copper dyshomeostasis in devastating pathogenesis in neurodegenerative diseases. Finally, we explore multiple unsolved questions and highlight the existing challenges to understand how copper homeostasis is modulated through membrane trafficking.
Collapse
Affiliation(s)
- Meng-Hsuan Wen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Xihong Xie
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Pei-San Huang
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Karen Yang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Tai-Yen Chen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
30
|
Sadia K, Ashraf MZ, Mishra A. Therapeutic Role of Sirtuins Targeting Unfolded Protein Response, Coagulation, and Inflammation in Hypoxia-Induced Thrombosis. Front Physiol 2021; 12:733453. [PMID: 34803727 PMCID: PMC8602789 DOI: 10.3389/fphys.2021.733453] [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: 07/15/2021] [Accepted: 10/19/2021] [Indexed: 12/23/2022] Open
Abstract
Thrombosis remains one of the leading causes of morbidity and mortality across the world. Many pathological milieus in the body resulting from multiple risk factors escort thrombosis. Hypoxic condition is one such risk factor that disturbs the integrity of endothelial cells to cause an imbalance between anticoagulant and procoagulant proteins. Hypoxia generates reactive oxygen species (ROS) and triggers inflammatory pathways to augment the coagulation cascade. Hypoxia in cells also activates unfolded protein response (UPR) signaling pathways in the endoplasmic reticulum (ER), which tries to restore ER homeostasis and function. But the sustained UPR linked with inflammation, generation of ROS and apoptosis stimulates the severity of thrombosis in the body. Sirtuins, a group of seven proteins, play a vast role in bringing down inflammation, oxidative and ER stress and apoptosis. As a result, sirtuins might provide a therapeutic approach towards the treatment or prevention of hypoxia-induced thrombosis. Sirtuins modulate hypoxia-inducible factors (HIFs) and counteract ER stress-induced apoptosis by attenuating protein kinase RNA-like endoplasmic reticulum kinase (PERK)/Eukaryotic translation initiation factor 2α (eIF2α) pathway activation. It prevents ER-stress mediated inflammation by targeting X-Box Binding Protein 1 (XBP1) and inhibiting nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κβ) signaling through deacetylation. Sirtuins also obstruct nucleotide-binding domain, leucine-rich-containing family, pyrin domain containing 3 (NLRP3) inflammasome activation to reduce the expression of several pro-inflammatory molecules. It protects cells against oxidative stress by targeting nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione (GSH), forkhead box O3 (FOXO3), superoxide dismutase (SOD), catalase (CAT), peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), glucose-6-phosphate dehydrogenase (G6PD), phosphoglucomutase-2 (PGAM2), and NF-κB, to name few. This review, thus, discusses the potential role of sirtuins as a new treatment for hypoxia-induced thrombosis that involves an intersection of UPR and inflammatory pathways in its pathological manifestation.
Collapse
Affiliation(s)
- Khan Sadia
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | | | - Aastha Mishra
- Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India
| |
Collapse
|
31
|
Morro B, Broughton R, Balseiro P, Handeland SO, Mackenzie S, Doherty MK, Whitfield PD, Shimizu M, Gorissen M, Sveier H, Albalat A. Endoplasmic reticulum stress as a key mechanism in stunted growth of seawater rainbow trout (Oncorhynchus mykiss). BMC Genomics 2021; 22:824. [PMID: 34781893 PMCID: PMC8594166 DOI: 10.1186/s12864-021-08153-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/01/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Rainbow trout (Oncorhynchus mykiss) is a salmonid species with a complex life-history. Wild populations are naturally divided into freshwater residents and sea-run migrants. Migrants undergo an energy-demanding adaptation for life in seawater, known as smoltification, while freshwater residents display these changes in an attenuated magnitude and rate. Despite this, in seawater rainbow trout farming all fish are transferred to seawater. Under these circumstances, weeks after seawater transfer, a significant portion of the fish die (around 10%) or experience growth stunting (GS; around 10%), which represents an important profitability and welfare issue. The underlying causes leading to GS in seawater-transferred rainbow trout remain unknown. In this study, we aimed at characterising the GS phenotype in seawater-transferred rainbow trout using untargeted and targeted approaches. To this end, the liver proteome (LC-MS/MS) and lipidome (LC-MS) of GS and fast-growing phenotypes were profiled to identify molecules and processes that are characteristic of the GS phenotype. Moreover, the transcription, abundance or activity of key proteins and hormones related to osmoregulation (Gill Na+, K + -ATPase activity), growth (plasma IGF-I, and liver igf1, igfbp1b, ghr1 and ctsl) and stress (plasma cortisol) were measured using targeted approaches. RESULTS No differences in Gill Na+, K + -ATPase activity and plasma cortisol were detected between the two groups. However, a significant downregulation in plasma IGF-I and liver igf1 transcription pointed at this growth factor as an important pathomechanism for GS. Changes in the liver proteome revealed reactive-oxygen-species-mediated endoplasmic reticulum stress as a key mechanism underlying the GS phenotype. From the lipidomic analysis, key observations include a reduction in triacylglycerols and elevated amounts of cardiolipins, a characteristic lipid class associated with oxidative stress, in GS phenotype. CONCLUSION While the triggers to the activation of endoplasmic reticulum stress are still unknown, data from this study point towards a nutritional deficiency as an underlying driver of this phenotype.
Collapse
Affiliation(s)
- Bernat Morro
- Institute of Aquaculture, University of Stirling, Stirling, UK
| | | | - Pablo Balseiro
- NORCE AS, Bergen, Norway.,Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Sigurd O Handeland
- NORCE AS, Bergen, Norway.,Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Simon Mackenzie
- Institute of Aquaculture, University of Stirling, Stirling, UK.,NORCE AS, Bergen, Norway
| | - Mary K Doherty
- Institute of Health Research and Innovation, Centre for Health Science, University of the Highlands and Islands, Scotland, UK
| | - Phillip D Whitfield
- Institute of Health Research and Innovation, Centre for Health Science, University of the Highlands and Islands, Scotland, UK.,Institute of Infection, Immunity and Inflammation, University of Glasgow, Scotland, UK
| | - Munetaka Shimizu
- Faculty of Fisheries Sciences, Hokkaido University, Sapporo, Japan
| | - Marnix Gorissen
- Department of Animal Ecology and Physiology, Radboud University, Institute of Water and Wetland Research, Nijmegen, The Netherlands
| | | | - Amaya Albalat
- Institute of Aquaculture, University of Stirling, Stirling, UK.
| |
Collapse
|
32
|
Demasi M, Augusto O, Bechara EJH, Bicev RN, Cerqueira FM, da Cunha FM, Denicola A, Gomes F, Miyamoto S, Netto LES, Randall LM, Stevani CV, Thomson L. Oxidative Modification of Proteins: From Damage to Catalysis, Signaling, and Beyond. Antioxid Redox Signal 2021; 35:1016-1080. [PMID: 33726509 DOI: 10.1089/ars.2020.8176] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.
Collapse
Affiliation(s)
- Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Etelvino J H Bechara
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Renata N Bicev
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Cerqueira
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | - Fernanda M da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Denicola
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lía M Randall
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Cassius V Stevani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Leonor Thomson
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
33
|
Mahmood F, Xu R, Awan MUN, Song Y, Han Q, Xia X, Zhang J. PDIA3: Structure, functions and its potential role in viral infections. Biomed Pharmacother 2021; 143:112110. [PMID: 34474345 DOI: 10.1016/j.biopha.2021.112110] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 02/08/2023] Open
Abstract
The catalysis of disulphide (SS) bonds is the most important characteristic of protein disulphide isomerase (PDI) family. Catalysis occurs in the endoplasmic reticulum, which contains many proteins, most of which are secretory in nature and that have at least one s-s bond. Protein disulphide isomerase A3 (PDIA3) is a member of the PDI family that acts as a chaperone. PDIA3 is highly expressed in response to cellular stress, and also intercept the apoptotic cellular death related to endoplasmic reticulum (ER) stress, and protein misfolding. PDIA3 expression is elevated in almost 70% of cancers and its expression has been linked with overall low cell invasiveness, survival and metastasis. Viral diseases present a significant public health threat. The presence of PDIA3 on the cell surface helps different viruses to enter the cells and also helps in replication. Therefore, inhibitors of PDIA3 have great potential to interfere with viral infections. In this review, we summarize what is known about the basic structure, functions and role of PDIA3 in viral infections. The review will inspire studies of pathogenic mechanisms and drug targeting to counter viral diseases.
Collapse
Affiliation(s)
- Faisal Mahmood
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Ruixian Xu
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Maher Un Nisa Awan
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Yuzhu Song
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Qinqin Han
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Xueshan Xia
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China.
| | - Jinyang Zhang
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China.
| |
Collapse
|
34
|
Gong FX, Zhan G, Han R, Yang Z, Fu X, Xiao R. De-dimerization of PTB is catalyzed by PDI and is involved in the regulation of p53 translation. Nucleic Acids Res 2021; 49:9342-9352. [PMID: 34403458 PMCID: PMC8450096 DOI: 10.1093/nar/gkab708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 12/03/2022] Open
Abstract
Polypyrimidine tract-binding protein (PTB) is an RNA binding protein existing both as dimer and monomer and shuttling between nucleus and cytoplasm. However, the regulation of PTB dimerization and the relationship between their functions and subcellular localization are unknown. Here we find that PTB presents as dimer and monomer in nucleus and cytoplasm respectively, and a disulfide bond involving Cysteine 23 is critical for the dimerization of PTB. Additionally, protein disulfide isomerase (PDI) is identified to be the enzyme that catalyzes the de-dimerization of PTB, which is dependent on the CGHC active site of the a’ domain of PDI. Furthermore, upon DNA damage induced by topoisomerase inhibitors, PTB is demonstrated to be de-dimerized with cytoplasmic accumulation. Finally, cytoplasmic PTB is found to associate with the ribosome and enhances the translation of p53. Collectively, these findings uncover a previously unrecognized mechanism of PTB dimerization, and shed light on the de-dimerization of PTB functionally linking to cytoplasmic localization and translational regulation.
Collapse
Affiliation(s)
- Fu-Xing Gong
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Guoqin Zhan
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Rong Han
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Zhigang Yang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| |
Collapse
|
35
|
Meng J, Wang L, Wang C, Zhao G, Wang H, Xu B, Guo X. AccPDIA6 from Apis cerana cerana plays important roles in antioxidation. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2021; 175:104830. [PMID: 33993956 DOI: 10.1016/j.pestbp.2021.104830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/06/2021] [Accepted: 03/13/2021] [Indexed: 06/12/2023]
Abstract
PDIA6 is a member of the protein disulfide isomerase (PDI) family, shows disulfide isomerase activity and oxidoreductase activity, and can act as a molecular chaperone. Its biological functions include modulating apoptosis, regulating the proliferation and invasion of cancer cells, supporting thrombosis and modulating insulin secretion. However, the roles of PDIA6 in Apis cerana cerana are poorly understood. Herein, we obtained the PDIA6 gene from A. cerana cerana (AccPDIA6). We investigated the expression patterns of AccPDIA6 in response to oxidative stress induced by H2O2, UV, HgCl2, extreme temperatures (4 °C, 42 °C) and pesticides (thiamethoxam and hexythiazox) and found that AccPDIA6 was upregulated by these treatments. Western blot analysis indicated that AccPDIA6 was also upregulated by oxidative stress at the protein level. In addition, a survival test demonstrated that the survival rate of E. coli cells expressing AccPDIA6 increased under oxidative stress, suggesting a possible antioxidant function of AccPDIA6. In addition, we tested the transcripts of other antioxidant genes and found that some of them were downregulated in AccPDIA6 knockdown samples. It was also discovered that the antioxidant enzymatic activity of superoxide dismutase (SOD) decreased in AccPDIA6-silenced bees. Moreover, the survival rate of AccPDIA6 knockdown bees decreased under oxidative stress, implying that AccPDIA6 may function in the oxidative stress response by enhancing the viability of honeybees. Taken together, these results indicated that AccPDIA6 may play an essential role in counteracting oxidative stress.
Collapse
Affiliation(s)
- Jie Meng
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Lijun Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Chen Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Guangdong Zhao
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Hongfang Wang
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Baohua Xu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, PR China.
| | - Xingqi Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China.
| |
Collapse
|
36
|
Xu X, Chiu J, Chen S, Fang C. Pathophysiological roles of cell surface and extracellular protein disulfide isomerase and their molecular mechanisms. Br J Pharmacol 2021; 178:2911-2930. [PMID: 33837960 DOI: 10.1111/bph.15493] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 03/23/2021] [Accepted: 04/04/2021] [Indexed: 12/21/2022] Open
Abstract
Protein disulfide isomerase (PDI) is the prototypic member of the thiol isomerase family that catalyses disulfide bond rearrangement. Initially identified in the endoplasmic reticulum as folding catalysts, PDI and other members in its family have also been widely reported to reside on the cell surface and in the extracellular matrix. Although how PDI is exported and retained on the cell surface remains a subject of debate, this unique pool of PDI is developing into an important mechanism underlying the redox regulation of protein sulfhydryls that are critical for the cellular activities under various disease conditions. This review aims to provide an overview of the pathophysiological roles of surface and extracellular PDI and their underlying molecular mechanisms. Understanding the involvement of extracellular PDI in these diseases will advance our knowledge in the molecular aetiology to facilitate the development of novel pharmacological strategies by specifically targeting PDI in extracellular compartments.
Collapse
Affiliation(s)
- Xulin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Joyce Chiu
- The Centenary Institute, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Shuai Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| |
Collapse
|
37
|
Hasipek M, Grabowski D, Guan Y, Alugubelli RR, Tiwari AD, Gu X, DeAvila GA, Silva AS, Meads MB, Parker Y, Lindner DJ, Saunthararajah Y, Shain KH, Maciejewski JP, Reu FJ, Phillips JG, Jha BK. Therapeutic Targeting of Protein Disulfide Isomerase PDIA1 in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13112649. [PMID: 34071205 PMCID: PMC8198550 DOI: 10.3390/cancers13112649] [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: 04/27/2021] [Revised: 05/19/2021] [Accepted: 05/22/2021] [Indexed: 01/20/2023] Open
Abstract
Multiple myeloma is a genetically complex hematologic neoplasia in which malignant plasma cells constantly operate at the maximum limit of their unfolded protein response (UPR) due to a high secretory burden of immunoglobulins and cytokines. The endoplasmic reticulum (ER) resident protein disulfide isomerase, PDIA1 is indispensable for maintaining structural integrity of cysteine-rich antibodies and cytokines that require accurate intramolecular disulfide bond arrangement. PDIA1 expression analysis from RNA-seq of multiple myeloma patients demonstrated an inverse relationship with survival in relapsed or refractory disease, supporting its critical role in myeloma persistence. Using a structure-guided medicinal chemistry approach, we developed a potent, orally bioavailable small molecule PDIA1 inhibitor CCF642-34. The inhibition of PDIA1 overwhelms the UPR in myeloma cells, resulting in their apoptotic cell death at doses that do not affect the normal CD34+ hematopoietic stem and progenitor cells. Bortezomib resistance leads to increased PDIA1 expression and thus CCF642-34 sensitivity, suggesting that proteasome inhibitor resistance leads to PDIA1 dependence for proteostasis and survival. CCF642-34 induces acute unresolvable UPR in myeloma cells, and oral treatment increased survival of mice in the syngeneic 5TGM1 model of myeloma. Results support development of CCF642-34 to selectively target the plasma cell program and overcome the treatment-refractory state in myeloma.
Collapse
Affiliation(s)
- Metis Hasipek
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
| | - Dale Grabowski
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
| | - Yihong Guan
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
| | - Raghunandan Reddy Alugubelli
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (R.R.A.); (G.A.D.); (K.H.S.); (F.J.R.)
| | - Anand D. Tiwari
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
| | - Xiaorong Gu
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
| | - Gabriel A. DeAvila
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (R.R.A.); (G.A.D.); (K.H.S.); (F.J.R.)
| | - Ariosto S. Silva
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (A.S.S.); (M.B.M.)
| | - Mark B. Meads
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (A.S.S.); (M.B.M.)
| | - Yvonne Parker
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
| | - Daniel J. Lindner
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
| | - Yogen Saunthararajah
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
- Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Kenneth H. Shain
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (R.R.A.); (G.A.D.); (K.H.S.); (F.J.R.)
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
- Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Frederic J. Reu
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (R.R.A.); (G.A.D.); (K.H.S.); (F.J.R.)
| | - James G. Phillips
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44195, USA; (M.H.); (D.G.); (Y.G.); (A.D.T.); (X.G.); (Y.P.); (D.J.L.); (Y.S.); (J.P.M.); (J.G.P.)
- Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Correspondence: ; Tel.: +1-216-444-6739
| |
Collapse
|
38
|
Singh Y, Nair AM, Verma PK. Surviving the odds: From perception to survival of fungal phytopathogens under host-generated oxidative burst. PLANT COMMUNICATIONS 2021; 2:100142. [PMID: 34027389 PMCID: PMC8132124 DOI: 10.1016/j.xplc.2021.100142] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/04/2020] [Accepted: 01/01/2021] [Indexed: 05/04/2023]
Abstract
Fungal phytopathogens pose a serious threat to global crop production. Only a handful of strategies are available to combat these fungal infections, and the increasing incidence of fungicide resistance is making the situation worse. Hence, the molecular understanding of plant-fungus interactions remains a primary focus of plant pathology. One of the hallmarks of host-pathogen interactions is the overproduction of reactive oxygen species (ROS) as a plant defense mechanism, collectively termed the oxidative burst. In general, high accumulation of ROS restricts the growth of pathogenic organisms by causing localized cell death around the site of infection. To survive the oxidative burst and achieve successful host colonization, fungal phytopathogens employ intricate mechanisms for ROS perception, ROS neutralization, and protection from ROS-mediated damage. Together, these countermeasures maintain the physiological redox homeostasis that is essential for cell viability. In addition to intracellular antioxidant systems, phytopathogenic fungi also deploy interesting effector-mediated mechanisms for extracellular ROS modulation. This aspect of plant-pathogen interactions is significantly under-studied and provides enormous scope for future research. These adaptive responses, broadly categorized into "escape" and "exploitation" mechanisms, are poorly understood. In this review, we discuss the oxidative stress response of filamentous fungi, their perception signaling, and recent insights that provide a comprehensive understanding of the distinct survival mechanisms of fungal pathogens in response to the host-generated oxidative burst.
Collapse
Affiliation(s)
- Yeshveer Singh
- Plant Immunity Laboratory, National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Athira Mohandas Nair
- Plant Immunity Laboratory, National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Praveen Kumar Verma
- Plant Immunity Laboratory, National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi 110067, India
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
39
|
Harrison MC, Niño LMJ, Rodrigues MA, Ryll J, Flatt T, Oettler J, Bornberg-Bauer E. Gene Coexpression Network Reveals Highly Conserved, Well-Regulated Anti-Ageing Mechanisms in Old Ant Queens. Genome Biol Evol 2021; 13:6263858. [PMID: 33944936 PMCID: PMC8214412 DOI: 10.1093/gbe/evab093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
Evolutionary theories of ageing predict a reduction in selection efficiency with age, a so-called “selection shadow,” due to extrinsic mortality decreasing effective population size with age. Classic symptoms of ageing include a deterioration in transcriptional regulation and protein homeostasis. Understanding how ant queens defy the trade-off between fecundity and lifespan remains a major challenge for the evolutionary theory of ageing. It has often been discussed that the low extrinsic mortality of ant queens, that are generally well protected within the nest by workers and soldiers, should reduce the selection shadow acting on old queens. We tested this by comparing strength of selection acting on genes upregulated in young and old queens of the ant, Cardiocondyla obscurior. In support of a reduced selection shadow, we find old-biased genes to be under strong purifying selection. We also analyzed a gene coexpression network (GCN) with the aim to detect signs of ageing in the form of deteriorating regulation and proteostasis. We find no evidence for ageing. In fact, we detect higher connectivity in old queens indicating increased transcriptional regulation with age. Within the GCN, we discover five highly correlated modules that are upregulated with age. These old-biased modules regulate several antiageing mechanisms such as maintenance of proteostasis, transcriptional regulation, and stress response. We observe stronger purifying selection on central hub genes of these old-biased modules compared with young-biased modules. These results indicate a lack of transcriptional ageing in old C. obscurior queens, possibly facilitated by strong selection at old age and well-regulated antiageing mechanisms.
Collapse
Affiliation(s)
- Mark C Harrison
- Institute for Evolution and Biodiversity, University of Münster, Germany
| | | | | | - Judith Ryll
- Institute for Evolution and Biodiversity, University of Münster, Germany
| | - Thomas Flatt
- Department of Biology, University of Fribourg, Switzerland
| | - Jan Oettler
- Institut für Zoologie/Evolutionsbiologie, University of Regensburg, Germany
| | - Erich Bornberg-Bauer
- Department of Biology, University of Fribourg, Switzerland.,Department of Protein Evolution, Max Planck Institute for Developmental Biology, Tübingen, Germany
| |
Collapse
|
40
|
Yu XJ, Xin GR, Liu KL, Liu XJ, Fu LY, Qi J, Kang KB, Meng TT, Yi QY, Li Y, Sun YJ, Kang YM. Paraventricular Nucleus Infusion of Oligomeric Proantho Cyanidins Improves Renovascular Hypertension. Front Neurosci 2021; 15:642015. [PMID: 33746706 PMCID: PMC7969814 DOI: 10.3389/fnins.2021.642015] [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: 12/15/2020] [Accepted: 02/10/2021] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress plays an important role in the pathogenesis of hypertension. Oligomeric proantho cyanidins (OPC) is the main polyphenol presents in grape seed and is known for its potent antioxidant and anti-inflammatory properties. In the present study, we hypothesize that OPC can attenuate oxidative stress in the paraventricular nucleus of hypothalamus (PVN), ameliorate neurotransmitter imbalance, decrease the blood pressure and sympathetic activity in renovascular hypertensive rats. After induction of renovascular hypertension by the two-kidney one-clip (2K-1C) method, male Sprague-Dawley rats received chronic bilateral PVN infusion of OPC (20 μg/h) or vehicle via osmotic minipump for 4 weeks. We found that hypertension induced by 2K-1C was associated with the production of reactive oxygen species (ROS) in the PVN. Infusion of OPC in the PVN significantly reduced the systolic blood pressure and norepinephrine in plasma of 2K-1C rats. In addition, PVN infusion of OPC decreased the level of ROS and the expression of stress-related nicotinamide adenine dinucleotide phosphate (NADPH) oxidases subunit NOX4, increased the levels of nuclear factor E2-related factor 2 (Nrf2) and antioxidant enzyme, balanced the content of cytokines, increased expression of glutamic acid decarboxylase and decreased the expression of tyrosine hydroxylase in the PVN of 2K-1C rats. Our findings provided strong evidence that PVN infusion of OPC inhibited the progression of renovascular hypertension through its potent anti-oxidative and anti-inflammatory function in the PVN.
Collapse
Affiliation(s)
- Xiao-Jing Yu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an, China
| | - Guo-Rui Xin
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an, China
| | - Kai-Li Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an, China
| | - Xiao-Jing Liu
- Department of Cardiology, The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Li-Yan Fu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an, China
| | - Jie Qi
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an, China
| | - Kai B Kang
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Ting-Ting Meng
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an, China
| | - Qiu-Yue Yi
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University, Xi'an, China
| | - Ying Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an, China
| | - Yao-Jun Sun
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an, China
| |
Collapse
|
41
|
Al-Fadhli FM, Afqi M, Sairafi MH, Almuntashri M, Alharby E, Alharbi G, Abdud Samad F, Hashmi JA, Zaytuni D, Bahashwan AA, Choi JH, Peake RWA, Beutler B, Almontashiri NAM. Biallelic loss of function variant in the unfolded protein response gene PDIA6 is associated with asphyxiating thoracic dystrophy and neonatal-onset diabetes. Clin Genet 2021; 99:694-703. [PMID: 33495992 DOI: 10.1111/cge.13930] [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: 12/19/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 12/18/2022]
Abstract
Protein disulfide isomerase A6 (PDIA6) is an unfolded protein response (UPR)-regulating protein. PDIA6 regulates the UPR sensing proteins, Inositol requiring enzyme 1, and EIF2AK3. Biallelic inactivation of the two genes in mice and humans resulted in embryonic lethality, diabetes, skeletal defects, and renal insufficiency. We recently showed that PDIA6 inactivation in mice caused embryonic and early lethality, diabetes and immunodeficiency. Here, we present a case with asphyxiating thoracic dystrophy (ATD) syndrome and infantile-onset diabetes. Whole exome sequencing revealed a homozygous frameshift variant in the PDIA6 gene. RNA expression was reduced in a gene dosage-dependent manner, supporting a loss-of-function effect of this variant. Phenotypic correlation with the mouse model recapitulated the growth defect and delay, early lethality, coagulation, diabetes, immunological, and polycystic kidney disease phenotypes. In general, the phenotype of the current patient is consistent with phenotypes associated with the disruption of PDIA6 and the sensors of UPR in mice and humans. This is the first study to associate ATD to the UPR gene, PDIA6. We recommend screening ATD cases with or without insulin-dependent diabetes for variants in PDIA6.
Collapse
Affiliation(s)
- Fatima M Al-Fadhli
- Unit of Genetic Diseases, Department of Pediatrics, Maternity and Children's Hospital, Almadinah Almunwarah, Saudi Arabia
| | - Manal Afqi
- Unit of Genetic Diseases, Department of Pediatrics, Maternity and Children's Hospital, Almadinah Almunwarah, Saudi Arabia
| | - Mona Hamza Sairafi
- Department of Nephrology, Maternity and Children's Hospital, Almadinah Almunwarah, Saudi Arabia
| | - Makki Almuntashri
- Department of Radiology, King Abdulaziz Medical City, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Essa Alharby
- Center for Genetics and Inherited Diseases, Taibah University, Almadinah Almunwarah, Saudi Arabia
| | - Ghadeer Alharbi
- Center for Genetics and Inherited Diseases, Taibah University, Almadinah Almunwarah, Saudi Arabia
| | - Firoz Abdud Samad
- Center for Genetics and Inherited Diseases, Taibah University, Almadinah Almunwarah, Saudi Arabia
| | - Jamil Amjad Hashmi
- Center for Genetics and Inherited Diseases, Taibah University, Almadinah Almunwarah, Saudi Arabia
| | - Dimah Zaytuni
- Center for Genetics and Inherited Diseases, Taibah University, Almadinah Almunwarah, Saudi Arabia
| | - Ahmed A Bahashwan
- Laboratory and Blood Bank Department, Maternity and Children's Hospital, Almadinah Almunwarah, Saudi Arabia
| | - Jin Huk Choi
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Roy W A Peake
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Naif A M Almontashiri
- Center for Genetics and Inherited Diseases, Taibah University, Almadinah Almunwarah, Saudi Arabia.,Faculty of Applied Medical Sciences, Taibah University, Almadinah Almunwarah, Saudi Arabia
| |
Collapse
|
42
|
Vona R, Pallotta L, Cappelletti M, Severi C, Matarrese P. The Impact of Oxidative Stress in Human Pathology: Focus on Gastrointestinal Disorders. Antioxidants (Basel) 2021; 10:201. [PMID: 33573222 PMCID: PMC7910878 DOI: 10.3390/antiox10020201] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidence shows that oxidative stress plays an essential role in the pathogenesis and progression of many diseases. The imbalance between the production of reactive oxygen species (ROS) and the antioxidant systems has been extensively studied in pulmonary, neurodegenerative cardiovascular disorders; however, its contribution is still debated in gastrointestinal disorders. Evidence suggests that oxidative stress affects gastrointestinal motility in obesity, and post-infectious disorders by favoring the smooth muscle phenotypic switch toward a synthetic phenotype. The aim of this review is to gain insight into the role played by oxidative stress in gastrointestinal pathologies (GIT), and the involvement of ROS in the signaling underlying the muscular alterations of the gastrointestinal tract (GIT). In addition, potential therapeutic strategies based on the use of antioxidants for the treatment of inflammatory gastrointestinal diseases are reviewed and discussed. Although substantial progress has been made in identifying new techniques capable of assessing the presence of oxidative stress in humans, the biochemical-molecular mechanisms underlying GIT mucosal disorders are not yet well defined. Therefore, further studies are needed to clarify the mechanisms through which oxidative stress-related signaling can contribute to the alteration of the GIT mucosa in order to devise effective preventive and curative therapeutic strategies.
Collapse
Affiliation(s)
- Rosa Vona
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Lucia Pallotta
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (L.P.); (M.C.); (C.S.)
| | - Martina Cappelletti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (L.P.); (M.C.); (C.S.)
| | - Carola Severi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (L.P.); (M.C.); (C.S.)
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
43
|
Biochemical pathways of copper complexes: progress over the past 5 years. Drug Discov Today 2021; 26:1086-1096. [PMID: 33486113 DOI: 10.1016/j.drudis.2021.01.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/21/2022]
Abstract
Copper is an essential trace element with vital roles in many metalloenzymes; it is also prominent among nonplatinum anticancer metallodrugs. Copper-based complexes are endogenously biocompatible, tenfold more potent than cisplatin, exhibit fewer adverse effects, and have a wide therapeutic window. In cancer biology, copper acts as an antitumor agent by inhibiting cancer via multiple pathways. Herein, we present an overview of advances in copper complexes as 'lead' antitumor drug candidates, and in understanding their biochemical and pharmacological pathways over the past 5 years. This review will help to develop more efficacious therapeutics to improve clinical outcomes for cancer treatments.
Collapse
|
44
|
Ferreira MKM, Aragão WAB, Bittencourt LO, Puty B, Dionizio A, Souza MPCD, Buzalaf MAR, de Oliveira EH, Crespo-Lopez ME, Lima RR. Fluoride exposure during pregnancy and lactation triggers oxidative stress and molecular changes in hippocampus of offspring rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111437. [PMID: 33096359 DOI: 10.1016/j.ecoenv.2020.111437] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 05/28/2023]
Abstract
Long-term exposure to high concentrations of fluoride (F) can damage mineralized and soft tissues such as bones, liver, kidney, intestine, and nervous system of adult rats. The high permeability of the blood-brain barrier and placenta to F during pregnancy and lactation may be critical to neurological development. Therefore, this study aimed to investigate the effects of F exposure during pregnancy and lactation on molecular processes and oxidative biochemistry of offspring rats' hippocampus. Pregnant Wistar rats were randomly assigned into 3 groups in accordance with the drinking water received: G1 - deionized water (control); G2 - 10 mg/L of F and G3 - 50 mg/L of F. The exposure to fluoridated water began on the first day of pregnancy and lasted until the 21st day of breastfeeding (when the offspring rats were weaned). Blood plasma samples of the offspring rats were collected to determine F levels. Hippocampi samples were collected for oxidative biochemistry analyses through antioxidant capacity against peroxyl (ACAP), lipid peroxidation (LPO), and nitrite (NO2-) levels. Also, brain-derived neurotrophic factor (BDNF) gene expression (RT-qPCR) and proteomic profile analyses were performed. The results showed that exposure to both F concentrations during pregnancy and lactation increased the F bioavailability, triggered redox imbalance featured by a decrease of ACAP, increase of LPO and NO2- levels, BDNF overexpression and changes in the hippocampus proteome. These findings raise novel questions regarding potential repercussions on the hippocampus structure and functioning in the different cognitive domains.
Collapse
Affiliation(s)
- Maria Karolina Martins Ferreira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Walessa Alana Bragança Aragão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Bruna Puty
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Aline Dionizio
- Department of Biological Sciences, Bauru Dental School, University of São Paulo, Bauru, São Paulo, Brazil
| | | | | | | | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil.
| |
Collapse
|
45
|
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
|
46
|
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
|
47
|
Alhammad R, Khunchai S, Tongmuang N, Limjindaporn T, Yenchitsomanus PT, Mutti L, Krstic-Demonacos M, Demonacos C. Protein disulfide isomerase A1 regulates breast cancer cell immunorecognition in a manner dependent on redox state. Oncol Rep 2020; 44:2406-2418. [PMID: 33125139 PMCID: PMC7610313 DOI: 10.3892/or.2020.7816] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/01/2020] [Indexed: 12/21/2022] Open
Abstract
Oxidoreductase protein disulphide isomerases (PDI) are involved in the regulation of a variety of biological processes including the modulation of endoplasmic reticulum (ER) stress, unfolded protein response (UPR), ER-mitochondria communication and the balance between pro-survival and pro-death pathways. In the current study the role of the PDIA1 family member in breast carcinogenesis was investigated by measuring ROS generation, mitochondrial membrane disruption, ATP production and HLA-G protein levels on the surface of the cellular membrane in the presence or absence of PDIA1. The results showed that this enzyme exerted pro-apoptotic effects in estrogen receptor (ERα)-positive breast cancer MCF-7 and pro-survival in triple negative breast cancer (TNBC) MDA-MB-231 cells. ATP generation was upregulated in PDIA1-silenced MCF-7 cells and downregulated in PDIA1-silenced MDA-MB-231 cells in a manner dependent on the cellular redox status. Furthermore, MCF-7 and MDA-MB-231 cells in the presence of PDIA1 expressed higher surface levels of the non-classical human leukocyte antigen (HLA-G) under oxidative stress conditions. Evaluation of the METABRIC datasets showed that low PDIA1 and high HLA-G mRNA expression levels correlated with longer survival in both ERα-positive and ERα-negative stage 2 breast cancer patients. In addition, analysis of the PDIA1 vs. the HLA-G mRNA ratio in the subgroup of the living stage 2 breast cancer patients exhibiting low PDIA1 and high HLA-G mRNA levels revealed that the longer the survival time of the ratio was high PDIA1 and low HLA-G mRNA and occurred predominantly in ERα-positive breast cancer patients whereas in the same subgroup of the ERα-negative breast cancer mainly this ratio was low PDIA1 and high HLA-G mRNA. Taken together these results provide evidence supporting the view that PDIA1 is linked to several hallmarks of breast cancer pathways including the process of antigen processing and presentation and tumor immunorecognition.
Collapse
Affiliation(s)
- Rashed Alhammad
- Faculty of Biology Medicine and Health, School of Health Sciences, Division of Pharmacy and Optometry, University of Manchester, Manchester M13 9PT, UK
| | - Sasiprapa Khunchai
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Nopprarat Tongmuang
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Thawornchai Limjindaporn
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Pa-Thai Yenchitsomanus
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | | | - Constantinos Demonacos
- Faculty of Biology Medicine and Health, School of Health Sciences, Division of Pharmacy and Optometry, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
48
|
Stojak M, Milczarek M, Kurpinska A, Suraj-Prazmowska J, Kaczara P, Wojnar-Lason K, Banach J, Stachowicz-Suhs M, Rossowska J, Kalviņš I, Wietrzyk J, Chlopicki S. Protein Disulphide Isomerase A1 Is Involved in the Regulation of Breast Cancer Cell Adhesion and Transmigration via Lung Microvascular Endothelial Cells. Cancers (Basel) 2020; 12:cancers12102850. [PMID: 33023153 PMCID: PMC7601413 DOI: 10.3390/cancers12102850] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer cell cross-talk with the host endothelium plays a crucial role in metastasis, but the underlying mechanisms are still not fully understood. We studied the involvement of protein disulphide isomerase A1 (PDIA1) in human breast cancer cell (MCF-7 and MDA-MB-231) adhesion and transendothelial migration. For comparison, the role of PDIA1 in proliferation, migration, cell cycle and apoptosis was also assessed. Pharmacological inhibitor, bepristat 2a and PDIA1 silencing were used to inhibit PDIA1. Inhibition of PDIA1 by bepristat 2a markedly decreased the adhesion of breast cancer cells to collagen type I, fibronectin and human lung microvascular endothelial cells. Transendothelial migration of breast cancer cells across the endothelial monolayer was also inhibited by bepristat 2a, an effect not associated with changes in ICAM-1 expression or changes in cellular bioenergetics. The silencing of PDIA1 produced less pronounced anti-adhesive effects. However, inhibiting extracellular free thiols by non-penetrating blocker p-chloromercuribenzene sulphonate substantially inhibited adhesion. Using a proteomic approach, we identified that β1 and α2 integrins were the most abundant among all integrins in breast cancer cells as well as in lung microvascular endothelial cells, suggesting that integrins could represent a target for PDIA1. In conclusion, extracellular PDIA1 plays a major role in regulating the adhesion of cancer cells and their transendothelial migration, in addition to regulating cell cycle and caspase 3/7 activation by intracellular PDIA1. PDIA1-dependent regulation of cancer-endothelial cell interactions involves disulphide exchange and most likely integrin activation but is not mediated by the regulation of ICAM-1 expression or changes in cellular bioenergetics in breast cancer or endothelial cells.
Collapse
Affiliation(s)
- Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
| | - Magdalena Milczarek
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.M.); (J.B.); (M.S.-S.); (J.R.)
| | - Anna Kurpinska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
| | - Joanna Suraj-Prazmowska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
| | - Patrycja Kaczara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
| | - Kamila Wojnar-Lason
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland
| | - Joanna Banach
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.M.); (J.B.); (M.S.-S.); (J.R.)
| | - Martyna Stachowicz-Suhs
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.M.); (J.B.); (M.S.-S.); (J.R.)
| | - Joanna Rossowska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.M.); (J.B.); (M.S.-S.); (J.R.)
| | - Ivars Kalviņš
- Laboratory of Carbofunctional Compounds, Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia;
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.M.); (J.B.); (M.S.-S.); (J.R.)
- Correspondence: (J.W.); (S.C.)
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland; (M.S.); (A.K.); (J.S.-P.); (P.K.); (K.W.-L.)
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland
- Correspondence: (J.W.); (S.C.)
| |
Collapse
|
49
|
Lénon M, Ke N, Szady C, Sakhtah H, Ren G, Manta B, Causey B, Berkmen M. Improved production of Humira antibody in the genetically engineered Escherichia coli SHuffle, by co-expression of human PDI-GPx7 fusions. Appl Microbiol Biotechnol 2020; 104:9693-9706. [PMID: 32997203 PMCID: PMC7595990 DOI: 10.1007/s00253-020-10920-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022]
Abstract
Abstract Microbial production of antibodies offers the promise of cheap, fast, and efficient production of antibodies at an industrial scale. Limiting this capacity in prokaryotes is the absence of the post-translational machinery, present in dedicated antibody producing eukaryotic cell lines, such as B cells. There has been few and limited success in producing full-length, correctly folded, and assembled IgG in the cytoplasm of prokaryotic cell lines. One such success was achieved by utilizing the genetically engineered Escherichia coli strain SHuffle with an oxidative cytoplasm. Due to the genetic disruption of reductive pathways, SHuffle cells are under constant oxidative stress, including increased levels of hydrogen peroxide (H2O2). The oxidizing capacity of H2O2 was linked to improved disulfide bond formation, by expressing a fusion of two endoplasmic reticulum-resident proteins, the thiol peroxidase GPx7 and the protein disulfide isomerase, PDI. In concert, these proteins mediate disulfide transfer from H2O2 to target proteins via PDI-Gpx7 fusions. The potential of this new strain was tested with Humira, a blockbuster antibody usually produced in eukaryotic cells. Expression results demonstrate that the new engineered SHuffle strain (SHuffle2) could produce Humira IgG four-fold better than the parental strain, both in shake-flask and in high-density fermentation. These preliminary studies guide the field in genetically engineering eukaryotic redox pathways in prokaryotes for the production of complex macromolecules. Key points • A eukaryotic redox pathway was engineered into the E. coli strain SHuffle in order to improve the yield of the blockbuster antibody Humira. • The best peroxidase-PDI fusion was selected using bioinformatics and in vivo studies. • Improved yields of Humira were demonstrated at shake-flask and high-density fermenters. Electronic supplementary material The online version of this article (10.1007/s00253-020-10920-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marine Lénon
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
- Department of Microbiology, Stress Adaptation and Metabolism in Enterobacteria Unit, UMR CNRS 2001, Institut Pasteur, 25-28 Rue du Dr Roux, 75015, Paris, France
| | - Na Ke
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Cecily Szady
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Hassan Sakhtah
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
- Boston Institute of Biotechnology, LLC., Upstream Process Development, 225 Turnpike Road, Southborough, MA, 01772, USA
| | - Guoping Ren
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Bruno Manta
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
- Facultad de Medicina, Departamento de Bioquímica and Centro de Investigaciones Biomédicas, Universidad de la República, CP 11800, Montevideo, Uruguay
| | - Bryce Causey
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Mehmet Berkmen
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA.
| |
Collapse
|
50
|
Role of Endoplasmic Reticulum Stress in Atherosclerosis and Its Potential as a Therapeutic Target. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9270107. [PMID: 32963706 PMCID: PMC7499294 DOI: 10.1155/2020/9270107] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/29/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022]
Abstract
Endoplasmic reticulum (ER) stress is closely associated with atherosclerosis and related cardiovascular diseases (CVDs). It occurs due to various pathological factors that interfere with ER homeostasis, resulting in the accumulation of unfolded or misfolded proteins in the ER lumen, thereby causing ER dysfunction. Here, we discuss the role of ER stress in different types of cells in atherosclerotic lesions. This discussion includes the activation of apoptotic and inflammatory pathways induced by prolonged ER stress, especially in advanced lesional macrophages and endothelial cells (ECs), as well as common atherosclerosis-related ER stressors in different lesional cells, which all contribute to the clinical progression of atherosclerosis. In view of the important role of ER stress and the unfolded protein response (UPR) signaling pathways in atherosclerosis and CVDs, targeting these processes to reduce ER stress may be a novel therapeutic strategy.
Collapse
|