1
|
Faraji N, Ebadpour N, Abavisani M, Gorji A. Unlocking Hope: Therapeutic Advances and Approaches in Modulating the Wnt Pathway for Neurodegenerative Diseases. Mol Neurobiol 2025; 62:3630-3652. [PMID: 39313658 PMCID: PMC11790780 DOI: 10.1007/s12035-024-04462-4] [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: 04/04/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024]
Abstract
Neurodegenerative diseases (NDs) are conditions characterized by sensory, motor, and cognitive impairments due to alterations in the structure and function of neurons in the central nervous system (CNS). Despite their widespread occurrence, the exact causes of NDs remain largely elusive, and existing treatments fall short in efficacy. The Wnt signaling pathway is an emerging molecular pathway that has been linked to the development and progression of various NDs. Wnt signaling governs numerous cellular processes, such as survival, polarity, proliferation, differentiation, migration, and fate specification, via a complex network of proteins. In the adult CNS, Wnt signaling regulates synaptic transmission, plasticity, memory formation, neurogenesis, neuroprotection, and neuroinflammation, all essential for maintaining neuronal function and integrity. Dysregulation of both canonical and non-canonical Wnt signaling pathways contributes to neurodegeneration through various mechanisms, such as amyloid-β accumulation, tau protein hyperphosphorylation, dopaminergic neuron degeneration, and synaptic dysfunction, prompting investigations into Wnt modulation as a therapeutic target to restore neuronal function and prevent or delay neurodegenerative processes. Modulating Wnt signaling has the potential to restore neuronal function and impede or postpone neurodegenerative processes, offering a therapeutic approach for targeting NDs. In this article, the current knowledge about how Wnt signaling works in Alzheimer's disease and Parkinson's disease is discussed. Our study aims to explore the molecular mechanisms, recent discoveries, and challenges involved in developing Wnt-based therapies.
Collapse
Affiliation(s)
- Navid Faraji
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Epilepsy Research Center, Münster University, Münster, Germany.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Neurosurgery Department, Münster University, Münster, Germany.
| |
Collapse
|
2
|
Ueland T, Butt N, Lekva T, Ørn S, Manhenke C, Aukrust P, Larsen AI. High dose statin treatment reduces circulating Dickkopf-1 following acute myocardial infarction. Int J Cardiol 2024; 406:132035. [PMID: 38604450 DOI: 10.1016/j.ijcard.2024.132035] [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: 10/19/2023] [Revised: 03/25/2024] [Accepted: 04/07/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Secreted glycoproteins of the Dickkopf (DKK) family modify Wnt signaling and may influence plaque destabilization but their modulation by statins in MI patients is not known. METHODS We measured plasma DKK-1 and DKK-3 in patients with acute ST-segment elevation MI (STEMI) before percutaneous coronary intervention (PCI) and after 2 and 7 days and 2 months in patients receiving short-term high-dose (40 mg rosuvastatin, given before PCI; n = 25) and moderate dose (20 mg simvastatin, given the day after PCI; n = 34). In vitro modulation of DKK-1 in human umbilical vein endothelial cells (HUVECs) by statins were assessed. RESULTS (i) Patients receiving high dose rosuvastatin had a marked decline in DKK-1 at day 2 which was maintained throughout the study period. However, a more prevalent use of β-blockers in the simvastatin group, that could have contributed to higher DKK-1 levels in these patients. (ii) There was a strong correlation between baseline DKK-1 levels and change in DKK-1 from baseline to day 2 in patients receiving high dose rosuvastatin treatment. (iii) DKK-3 increased at day 2 but returned to baseline levels at 2 months in both treatment groups. (iv) Statin treatment dose-dependently decreased DKK-1 mRNA and protein levels in HUVEC. CONCLUSIONS Our findings suggest that high dose statin treatment with 40 mg rosuvastatin could persistently down-regulate DKK-1 levels, even at 2 months after the initial event in STEMI patients.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway.
| | - Noreen Butt
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | - Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Stein Ørn
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway; Department of Electrical Engineering and Computer Science, University of Stavanger, Stavanger, Norway
| | - Cord Manhenke
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Alf Inge Larsen
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway; Department of Clinical Science, University of Bergen, Norway
| |
Collapse
|
3
|
Liu X, Zheng T, Zhang Y, Zhao Y, Liu F, Dai S, Zhang M, Zhang W, Zhang C, Zhang M, Li X. Endothelial Dickkopf-1 Promotes Smooth Muscle Cell-derived Foam Cell Formation via USP53-mediated Deubiquitination of SR-A During Atherosclerosis. Int J Biol Sci 2024; 20:2943-2964. [PMID: 38904030 PMCID: PMC11186357 DOI: 10.7150/ijbs.91957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/27/2024] [Indexed: 06/22/2024] Open
Abstract
Background: Shear stress-induced Dickkopf-1 (DKK1) secretion by endothelial cells (ECs) promotes EC dysfunction and accelerates atherosclerosis (AS). However, the paracrine role of endothelial DKK1 in modulating adjacent smooth muscle cells (SMCs) in atherosclerosis remains unclear. This study investigated the role of EC-secreted DKK1 in SMC-derived foam cell formation under shear stress, in vitro and in vivo. Methods: Parallel-plate co-culture flow system was used to explore the cellular communication between ECs and SMCs under shear stress in vitro. Endothelium-specific knockout of DKK1 (DKK1ECKO/APOE-/-) and endothelium-specific overexpression of DKK1 (DKK1ECTg) mice were constructed to investigate the role of endothelial DKK1 in atherosclerosis and SMC-derived foam cell formation in vivo. RNA sequencing (RNA-seq) was used to identify the downstream targets of DKK1. Reverse transcription quantitative polymerase chain reaction (RT-qPCR), western blot, coimmunoprecipitation (Co-IP) assays and chromatin immunoprecipitation (ChIP) experiments were conducted to explore the underlying regulatory mechanisms. Results: DKK1 is transcriptionally upregulated in ECs under conditions of low shear stress, but not in co-cultured SMCs. However, DKK1 protein in co-cultured SMCs is increased via uptake of low shear stress-induced endothelial DKK1, thereby promoting lipid uptake and foam cell formation in co-cultured SMCs via the post-translational upregulation of scavenger receptor-A (SR-A) verified in parallel-plate co-culture flow system, DKK1ECKO and DKK1ECTg mice. RNA sequencing revealed that DKK1-induced SR-A upregulation in SMCs is dependent on Ubiquitin-specific Protease 53 (USP53), which bound to SR-A via its USP domain and cysteine at position 41, exerting deubiquitination to maintain the stability of the SR-A protein by removing the K48 ubiquitin chain and preventing proteasomal pathway degradation, thereby mediating the effect of DKK1 on lipid uptake in SMCs. Moreover, DKK1 regulates the transcription of USP53 by facilitating the binding of transcription factor CREB to the USP53 promoter. SMC-specific overexpression of USP53 via adeno-associated virus serotype 2 vectors in DKK1ECKO/APOE-/- mice reversed the alleviation of atherosclerotic plaque burden, SR-A expression and lipid accumulation in SMCs within plaques resulting from DKK1 deficiency. Conclusions: Our findings demonstrate that, endothelial DKK1, induced by pathological low shear stress, acts as an intercellular mediator, promoted the foam cell formation of SMCs. These results suggest that targeted intervention with endothelial DKK1 may confer beneficial effects on atherosclerosis.
Collapse
Affiliation(s)
- Xiaolin Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Tengfei Zheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yu Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yachao Zhao
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fengming Liu
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Shen Dai
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Meng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wencheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Mei Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
4
|
Piarulli F, Banfi C, Ragazzi E, Gianazza E, Munno M, Carollo M, Traldi P, Lapolla A, Sartore G. Multiplexed MRM-based proteomics for identification of circulating proteins as biomarkers of cardiovascular damage progression associated with diabetes mellitus. Cardiovasc Diabetol 2024; 23:36. [PMID: 38245742 PMCID: PMC10800045 DOI: 10.1186/s12933-024-02125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 01/07/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) increases the risk of coronary heart disease (CHD) by 2-4 fold, and is associated with endothelial dysfunction, dyslipidaemia, insulin resistance, and chronic hyperglycaemia. The aim of this investigation was to assess, by a multimarker mass spectrometry approach, the predictive role of circulating proteins as biomarkers of cardiovascular damage progression associated with diabetes mellitus. METHODS The study considered 34 patients with both T2DM and CHD, 31 patients with T2DM and without CHD, and 30 patients without diabetes with a diagnosis of CHD. Plasma samples of subjects were analysed through a multiplexed targeted liquid chromatography mass spectrometry (LC-MS)-based assay, namely Multiple Reaction Monitoring (MRM), allowing the simultaneous detection of peptides derived from a protein of interest. Gene Ontology (GO) Analysis was employed to identify enriched GO terms in the biological process, molecular function, or cellular component categories. Non-parametric multivariate methods were used to classify samples from patients and evaluate the relevance of the analysed proteins' panel. RESULTS A total of 81 proteins were successfully quantified in the human plasma samples. Gene Ontology analysis assessed terms related to blood microparticles, extracellular exosomes and collagen-containing extracellular matrix. Preliminary evaluation using analysis of variance (ANOVA) of the differences in the proteomic profile among patient groups identified 13 out of the 81 proteins as significantly different. Multivariate analysis, including cluster analysis and principal component analysis, identified relevant grouping of the 13 proteins. The first main cluster comprises apolipoprotein C-III, apolipoprotein C-II, apolipoprotein A-IV, retinol-binding protein 4, lysozyme C and cystatin-C; the second one includes, albeit with sub-grouping, alpha 2 macroglobulin, afamin, kininogen 1, vitronectin, vitamin K-dependent protein S, complement factor B and mannan-binding lectin serine protease 2. Receiver operating characteristic (ROC) curves obtained with the 13 selected proteins using a nominal logistic regression indicated a significant overall distinction (p < 0.001) among the three groups of subjects, with area under the ROC curve (AUC) ranging 0.91-0.97, and sensitivity and specificity ranging from 85 to 100%. CONCLUSIONS Targeted mass spectrometry approach indicated 13 multiple circulating proteins as possible biomarkers of cardiovascular damage progression associated with T2DM, with excellent classification results in terms of sensitivity and specificity.
Collapse
Affiliation(s)
| | - Cristina Banfi
- Centro Cardiologico Monzino, IRCCS, Milano, 20138, Italy.
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.
| | - Erica Gianazza
- Centro Cardiologico Monzino, IRCCS, Milano, 20138, Italy
| | - Marco Munno
- Centro Cardiologico Monzino, IRCCS, Milano, 20138, Italy
| | - Massimo Carollo
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Pietro Traldi
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | | | - Giovanni Sartore
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| |
Collapse
|
5
|
Li X, Zheng T, Zhang Y, Zhao Y, Liu F, Dai S, Liu X, Zhang M. Dickkopf-1 promotes vascular smooth muscle cell foam cell formation and atherosclerosis development through CYP4A11/SREBP2/ABCA1. FASEB J 2023; 37:e23048. [PMID: 37389895 DOI: 10.1096/fj.202300295r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/20/2023] [Accepted: 06/08/2023] [Indexed: 07/01/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are considered to be a crucial source of foam cells in atherosclerosis due to their low expression level of cholesterol exporter ATP-binding cassette transporter A1 (ABCA1) intrinsically. While the definite regulatory mechanisms are complicated and have not yet been fully elucidated, we previously reported that Dickkopf-1 (DKK1) mediates endothelial cell (EC) dysfunction, thereby aggravating atherosclerosis. However, the role of smooth muscle cell (SMC) DKK1 in atherosclerosis and foam cell formation remains unknown. In this study, we established SMC-specific DKK1-knockout (DKK1SMKO ) mice by crossbreeding DKK1flox/flox mice with TAGLN-Cre mice. Then, DKK1SMKO mice were crossed with APOE-/- mice to generate DKK1SMKO /APOE-/- mice, which exhibited milder atherosclerotic burden and fewer SMC foam cells. In vitro loss- and gain-of-function studies of DKK1 in primary human aortic smooth muscle cells (HASMCs) have proven that DKK1 prevented oxidized lipid-induced ABCA1 upregulation and cholesterol efflux and promoted SMC foam cell formation. Mechanistically, RNA-sequencing (RNA-seq) analysis of HASMCs as well as chromatin immunoprecipitation (ChIP) experiments showed that DKK1 mediates the binding of transcription factor CCAAT/enhancer-binding protein delta (C/EBPδ) to the promoter of cytochrome P450 epoxygenase 4A11 (CYP4A11) to regulate its expression. In addition, CYP4A11 as well as its metabolite 20-HETE-promoted activation of transcription factor sterol regulatory element-binding protein 2 (SREBP2) mediated the DKK1 regulation of ABCA1 in SMC. Furthermore, HET0016, the antagonist of CYP4A11, has also shown an alleviating effect on atherosclerosis. In conclusion, our results demonstrate that DKK1 promotes SMC foam cell formation during atherosclerosis via a reduction in CYP4A11-20-HETE/SREBP2-mediated ABCA1 expression.
Collapse
Affiliation(s)
- Xiao Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Tengfei Zheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yu Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yachao Zhao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fengming Liu
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Shen Dai
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiaolin Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Mei Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
6
|
Carrozzini T, Pollaci G, Gorla G, Potenza A, Rifino N, Acerbi F, Vetrano IG, Ferroli P, Bersano A, Gianazza E, Banfi C, Gatti L. Proteome Profiling of the Dura Mater in Patients with Moyamoya Angiopathy. Int J Mol Sci 2023; 24:11194. [PMID: 37446373 DOI: 10.3390/ijms241311194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Moyamoya angiopathy (MMA) is an uncommon cerebrovascular disease characterized by a progressive steno-occlusive lesion of the internal carotid artery and the compensatory development of an unstable network of collateral vessels. These vascular hallmarks are responsible for recurrent ischemic/hemorrhagic strokes. Surgical treatment represents the preferred procedure for MMA patients, and indirect revascularization may induce a spontaneous angiogenesis between the brain surface and dura mater (DM), whose function remains rather unknown. A better understanding of MMA pathogenesis is expected from the molecular characterization of DM. We performed a comprehensive, label-free, quantitative mass spectrometry-based proteomic characterization of DM. The 30 most abundant identified proteins were located in the extracellular region or exosomes and were involved in extracellular matrix organization. Gene ontology analysis revealed that most proteins were involved in binding functions and hydrolase activity. Among the 30 most abundant proteins, Filamin A is particularly relevant because considering its well-known biochemical functions and molecular features, it could be a possible second hit gene with a potential role in MMA pathogenesis. The current explorative study could pave the way for further analyses aimed at better understanding such uncommon and disabling intracranial vasculopathy.
Collapse
Affiliation(s)
- Tatiana Carrozzini
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Giuliana Pollaci
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, Università di Milano, 20133 Milan, Italy
| | - Gemma Gorla
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Antonella Potenza
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Nicola Rifino
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Francesco Acerbi
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
- Experimental Microsurgical Laboratory, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Ignazio G Vetrano
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
- Department of Biomedical Sciences for Health, Università di Milano, 20133 Milan, Italy
| | - Paolo Ferroli
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Anna Bersano
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Erica Gianazza
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Laura Gatti
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| |
Collapse
|
7
|
Prenylcysteine Oxidase 1 Is a Key Regulator of Adipogenesis. Antioxidants (Basel) 2023; 12:antiox12030542. [PMID: 36978789 PMCID: PMC10045348 DOI: 10.3390/antiox12030542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/18/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023] Open
Abstract
The process of adipogenesis involves the differentiation of preadipocytes into mature adipocytes. Excessive adipogenesis promotes obesity, a condition that increasingly threatens global health and contributes to the rapid rise of obesity-related diseases. We have recently shown that prenylcysteine oxidase 1 (PCYOX1) is a regulator of atherosclerosis-disease mechanisms, which acts through mechanisms not exclusively related to its pro-oxidant activity. To address the role of PCYOX1 in the adipogenic process, we extended our previous observations confirming that Pcyox1−/−/Apoe−/− mice fed a high-fat diet for 8 or 12 weeks showed significantly lower body weight, when compared to Pcyox1+/+/Apoe−/− mice, due to an evident reduction in visceral adipose content. We herein assessed the role of PCYOX1 in adipogenesis. Here, we found that PCYOX1 is expressed in adipose tissue, and, independently from its pro-oxidant enzymatic activity, is critical for adipogenesis. Pcyox1 gene silencing completely prevented the differentiation of 3T3-L1 preadipocytes, by acting as an upstream regulator of several key players, such as FABP4, PPARγ, C/EBPα. Proteomic analysis, performed by quantitative label-free mass spectrometry, further strengthened the role of PCYOX1 in adipogenesis by expanding the list of its downstream targets. Finally, the absence of Pcyox1 reduces the inflammatory markers in adipose tissue. These findings render PCYOX1 a novel adipogenic factor with possible pathophysiological or therapeutic potential.
Collapse
|
8
|
Pharmacometabolomics for the Study of Lipid-Lowering Therapies: Opportunities and Challenges. Int J Mol Sci 2023; 24:ijms24043291. [PMID: 36834701 PMCID: PMC9960554 DOI: 10.3390/ijms24043291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Lipid-lowering therapies are widely used to prevent the development of atherosclerotic cardiovascular disease (ASCVD) and related mortality worldwide. "Omics" technologies have been successfully applied in recent decades to investigate the mechanisms of action of these drugs, their pleiotropic effects, and their side effects, aiming to identify novel targets for future personalized medicine with an improvement of the efficacy and safety associated with the treatment. Pharmacometabolomics is a branch of metabolomics that is focused on the study of drug effects on metabolic pathways that are implicated in the variation of response to the treatment considering also the influences from a specific disease, environment, and concomitant pharmacological therapies. In this review, we summarized the most significant metabolomic studies on the effects of lipid-lowering therapies, including the most commonly used statins and fibrates to novel drugs or nutraceutical approaches. The integration of pharmacometabolomics data with the information obtained from the other "omics" approaches could help in the comprehension of the biological mechanisms underlying the use of lipid-lowering drugs in view of defining a precision medicine to improve the efficacy and reduce the side effects associated with the treatment.
Collapse
|
9
|
Xu H, Ding Z, Chen J, Zhang Y, Shan W, Chen X, Liu X, Gao Y, Han G. Correlation between serum Dickkopf-1 (DKK1) levels and coronary artery stenosis. Nutr Metab Cardiovasc Dis 2023; 33:168-176. [PMID: 36411225 DOI: 10.1016/j.numecd.2022.10.007] [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: 09/02/2021] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND AIMS To study the correlation between the level of serum Dickkopf-1 (DKK1) and the degree of coronary artery stenosis in patients with coronary atherosclerotic heart disease. METHODS AND RESULTS In 2018, general data and biochemical indexes of 311 patients who underwent coronary angiography were recorded. Before procedure, arterial blood was drawn and the concentrations of DKK1, retinol binding protein 4 (RBP4), plasminogen activator inhibitor (PAI-1) were measured. Based on coronary angiography results, subjects were divided into a coronary heart disease (CHD) group; and a non-coronary heart disease (non-CHD)group. The CHD group was divided into three subgroups: the low Gensini score; the middle Gensini score; and the high Gensini score subgroups. Compared with those of the non-CHD group, DKK1, RBP4 and PAI-1 of the CHD group were significantly higher, while the OC was lower. DKK1,RBP4 and PAI-1 levels of the middle and high Gensini subgroups were significantly higher, compared with that of the low Gensini subgroup. Differences between osteocalcin (OC), beta-isomerized C-terminal telopeptidase (β-CTX), and 25(OH)2D3 of the three subgroups were not significant. Correlation between DKK1 and the inflammatory factors, RBP4 and PAI-1, was positive. Correlation between DKK1 and β - CTX, 25(OH)2D3 and OC was not significant. DKK1 was a risk factor for CHD. The degree of coronary artery stenosis was related to DKK1 concentration. CONCLUSIONS Serum DKK1 levels in coronary heart disease patients were significantly higher, and positively correlated with the degree of coronary artery stenosis. DKK1 level is an independent risk factor for coronary heart disease.
Collapse
Affiliation(s)
- Hongxiu Xu
- Development of Endocrinology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, PR China; The First Hospital of Qinghuangdao, PR China
| | - Zhenjiang Ding
- Development of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, PR China
| | - Jiaoyue Chen
- Development of Endocrinology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, PR China
| | - Ying Zhang
- Development of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, PR China
| | - Weichao Shan
- Development of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, PR China
| | - Xiaoyu Chen
- Development of Endocrinology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, PR China
| | - Xiaoyan Liu
- Development of Endocrinology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, PR China
| | - Yu Gao
- Development of Endocrinology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, PR China
| | - Guiyan Han
- Development of Endocrinology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, PR China.
| |
Collapse
|
10
|
Mostafa Arabi S, Sadat Bahrami L, MalekAhmadi M, Chambari M, Milkarizi N, Orekhov AN, Sahebkar A. The effect of combination therapy with statins and ezetimibe on proinflammatory cytokines: A systematic review and meta-analysis of randomized controlled trials. Int Immunopharmacol 2022; 113:109477. [DOI: 10.1016/j.intimp.2022.109477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
|
11
|
Shimizu C, Kim J, He M, Tremoulet AH, Hoffman HM, Shyy JY, Burns JC. RNA Sequencing Reveals Beneficial Effects of Atorvastatin on Endothelial Cells in Acute Kawasaki Disease. J Am Heart Assoc 2022; 11:e025408. [DOI: 10.1161/jaha.122.025408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background
Damage to the coronary arteries during the acute phase of Kawasaki disease (KD) is linked to inflammatory cell infiltration, myointimal proliferation, and endothelial cell (EC) dysfunction. To understand the response of ECs to KD treatment, we studied the genome‐wide transcriptional changes in cultured ECs incubated with KD sera before and after treatment with or without atorvastatin.
Methods and Results
RNA sequencing of human umbilical vein ECs incubated with pooled sera from patients with acute KD before or after treatment with intravenous immunoglobulin and infliximab revealed differentially expressed genes in interleukin‐1, tumor necrosis factor‐α, and inflammatory cell recruitment pathways. Subacute sera pooled from patients treated with intravenous immunoglobulin, infliximab, and atorvastatin uniquely induced expression of
NOS3
, Kruppel like factor (
KLF2
, and
KLF4
(promotes EC homeostasis and angiogenesis) and ZFP36 ring finger protein (ZFP36) and suppressor of cytokine signaling 3 (SOCS3) (suppresses inflammation), and suppressed expression of
TGFB2
and
DKK1
(induces endothelial‐mesenchymal transition) and sphingosine kinase 1 (SPHK1) and C‐X‐C motif chemokine ligand 8 (CXCL8) (induces inflammation).
Conclusions
These results suggest that atorvastatin treatment of patients with acute KD may improve EC health, reduce mediators of inflammation produced by ECs, and block KD‐induced myofibroblast proliferation.
Collapse
Affiliation(s)
- Chisato Shimizu
- Department of Pediatrics University of California, San Diego La Jolla CA
| | - Jihoon Kim
- Department of Biomedical Informatics University of California, San Diego La Jolla CA
| | - Ming He
- Department of Medicine University of California, San Diego La Jolla CA
| | - Adriana H. Tremoulet
- Department of Pediatrics University of California, San Diego La Jolla CA
- Rady Children’s Hospital San Diego CA
| | - Hal M. Hoffman
- Department of Pediatrics University of California, San Diego La Jolla CA
- Rady Children’s Hospital San Diego CA
| | - John Y‐J. Shyy
- Department of Medicine University of California, San Diego La Jolla CA
| | - Jane C. Burns
- Department of Pediatrics University of California, San Diego La Jolla CA
- Rady Children’s Hospital San Diego CA
| |
Collapse
|
12
|
Tong XK, Royea J, Hamel E. Simvastatin rescues memory and granule cell maturation through the Wnt/β-catenin signaling pathway in a mouse model of Alzheimer's disease. Cell Death Dis 2022; 13:325. [PMID: 35397630 PMCID: PMC8994768 DOI: 10.1038/s41419-022-04784-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/10/2022] [Accepted: 03/22/2022] [Indexed: 12/25/2022]
Abstract
We previously showed that simvastatin (SV) restored memory in a mouse model of Alzheimer disease (AD) concomitantly with normalization in protein levels of memory-related immediate early genes in hippocampal CA1 neurons. Here, we investigated age-related changes in the hippocampal memory pathway, and whether the beneficial effects of SV could be related to enhanced neurogenesis and signaling in the Wnt/β-catenin pathway. APP mice and wild-type (WT) littermate controls showed comparable number of proliferating (Ki67-positive nuclei) and immature (doublecortin (DCX)-positive) granule cells in the dentate gyrus until 3 months of age. At 4 months, Ki67 or DCX positive cells decreased sharply and remained less numerous until the endpoint (6 months) in both SV-treated and untreated APP mice. In 6 month-old APP mice, dendritic extensions of DCX immature neurons in the molecular layer were shorter, a deficit fully normalized by SV. Similarly, whereas mature granule cells (calbindin-immunopositive) were decreased in APP mice and not restored by SV, their dendritic arborizations were normalized to control levels by SV treatment. SV increased Prox1 protein levels (↑67.7%, p < 0.01), a Wnt/β-catenin signaling target, while significantly decreasing (↓61.2%, p < 0.05) the upregulated levels of the β-catenin-dependent Wnt pathway inhibitor DKK1 seen in APP mice. In APP mice, SV benefits were recapitulated by treatment with the Wnt/β-catenin specific agonist WAY-262611, whereas they were fully abolished in mice that received the Wnt/β-catenin pathway inhibitor XAV939 during the last month of SV treatment. Our results indicate that activation of the Wnt-β-catenin pathway through downregulation of DKK1 underlies SV neuronal and cognitive benefits.
Collapse
Affiliation(s)
- Xin-Kang Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, H3A 2B4, Montréal, QC, Canada
| | - Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, H3A 2B4, Montréal, QC, Canada.,Department of Biochemistry, Microbiology, Immunology University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, H3A 2B4, Montréal, QC, Canada.
| |
Collapse
|
13
|
Seo SH, Kim E, Lee S, Lee Y, Han DH, Go H, Seong JK, Choi K. Inhibition of CXXC5 function reverses obesity-related metabolic diseases. Clin Transl Med 2022; 12:e742. [PMID: 35384342 PMCID: PMC8982507 DOI: 10.1002/ctm2.742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/28/2022] [Accepted: 02/07/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Metabolic diseases, including type 2 diabetes, have long been considered incurable, chronic conditions resulting from a variety of pathological conditions in obese patients. Growing evidence suggests the Wnt/β-catenin pathway is a major pathway in adipose tissue remodelling, pancreatic β-cell regeneration and energy expenditure through regulation of key metabolic target genes in various tissues. CXXC5-type zinc finger protein 5 (CXXC5) is identified negative feedback regulator of the Wnt/β-catenin pathway that functions via Dishevelled (Dvl) binding. METHODS Expression level of CXXC5 was characterised in clinical samples and diabetes-induced mice model. Diabetes-induced mice model was established by using high-fat diet (HFD). HFD-fed mice treated with KY19334, a small molecule inhibiting CXXC5-Dvl protein-protein interaction (PPI), was used to assess the role of CXXC5 in metabolic diseases. RESULTS Here, we show that CXXC5 is overexpressed with suppression of Wnt/β-catenin signalling in visceral adipose tissues of patients with obesity-related diabetes. Meanwhile, Cxxc5-/- mice fed an HFD exhibited resistance to metabolic dysregulation. KY19334 restores the lowered Wnt/β-catenin signalling and reverses metabolic abnormalities as observed in HFD-fed Cxxc5-/- mice. Administration of KY19334 on HFD-fed mice had a long-lasting glucose-controlling effect through remodelling of adipocytes and regeneration of pancreatic β-cells. CONCLUSION Overall, the inhibition of CXXC5 function by small molecule-mediated interference of Dvl binding is a potential therapeutic strategy for the treatment of obesity-related diabetes.
Collapse
Affiliation(s)
- Seol Hwa Seo
- Department of BiotechnologyCollege of Life Science and BiotechnologyYonsei UniversitySeoulRepublic of Korea
| | - Eunhwan Kim
- Department of BiotechnologyCollege of Life Science and BiotechnologyYonsei UniversitySeoulRepublic of Korea
| | | | - Yong‐ho Lee
- Department of Internal MedicineYonsei UniversitySeoulRepublic of Korea
| | - Dai Hoon Han
- Department of surgeryYonsei University College of MedicineSeoulRepublic of Korea
| | - Hyesun Go
- Korea Mouse Phenotyping CenterSeoul National UniversitySeoulRepublic of Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping CenterSeoul National UniversitySeoulRepublic of Korea
| | - Kang‐Yell Choi
- Department of BiotechnologyCollege of Life Science and BiotechnologyYonsei UniversitySeoulRepublic of Korea
- CK Regeon Inc.SeoulRepublic of Korea
| |
Collapse
|
14
|
Drug repurposing for stroke intervention. Drug Discov Today 2022; 27:1974-1982. [DOI: 10.1016/j.drudis.2022.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023]
|
15
|
Kasoha M, Takacs Z, Fackiner L, Gerlinger C, Sklavounos P, Radosa J, Solomayer EF, Hamza A. Comparison of Maternal Serum Levels and Placental mRNA Levels of Dickkopf-1 in Preeclamptic and Normal Pregnant Women at Delivery. Geburtshilfe Frauenheilkd 2021; 81:1247-1255. [PMID: 34754274 PMCID: PMC8568501 DOI: 10.1055/a-1557-1234] [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: 04/12/2021] [Accepted: 07/22/2021] [Indexed: 11/17/2022] Open
Abstract
Background
Preeclampsia remains a major cause of perinatal and maternal mortality and morbidity worldwide. Wnt/β-catenin signaling is known to be critically involved in placenta development processes. Dickkopf-1 (DKK1) is a key regulator of this transduction pathway. The aim of this study is to compare maternal serum DKK1 levels and placental mRNA levels of
DKK1
and β-catenin in preeclamptic and normal pregnant women at delivery.
Methods
The present study included 30 women with preeclampsia and 30 women with normal pregnancy. Maternal serum DKK1 levels were measured by ELISA. Placental mRNA levels of
DKK1
and β-catenin were detected using RT-PCR.
Results
Decreased maternal serum DKK1 levels were associated with worse maternal and fetal complications including HELLP syndrome, determination of one or more pathological symptom and IUGR diagnosis. No significant difference in maternal serum DKK1 levels was reported between preeclamptic women and women with normal pregnancy. Placental mRNA
DKK1
levels were lower in preeclamptic women compared with normal pregnant women. Placental mRNA β-catenin levels showed no significant difference between two groups.
Conclusions
Our findings reported the aberrant placental mRNA
DKK1
levels in patients with preeclampsia. In addition, worse preeclampsia features were associated with decreased maternal serum DKK1 levels. Hence, aberrant Wnt/β-catenin signaling might present a plausible mechanism in preeclampsia pathogenicity. Dysregulated expression of DKK1 at gene level in the placenta but not at protein level in the maternal serum might confirm the notion that preeclampsia is a type of placenta-derived disease.
Collapse
Affiliation(s)
- Mariz Kasoha
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, Homburg/Saar, Germany
| | - Zoltan Takacs
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, Homburg/Saar, Germany
| | - Lena Fackiner
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, Homburg/Saar, Germany
| | - Christoph Gerlinger
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, Homburg/Saar, Germany
| | - Panagiotis Sklavounos
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, Homburg/Saar, Germany
| | - Julia Radosa
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, Homburg/Saar, Germany
| | - Erich-Franz Solomayer
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, Homburg/Saar, Germany
| | - Amr Hamza
- Department of Gynecology, Obstetrics and Reproductive Medicine, University Medical School of Saarland, Homburg/Saar, Germany
| |
Collapse
|
16
|
Banfi C, Baetta R, Barbieri SS, Brioschi M, Guarino A, Ghilardi S, Sandrini L, Eligini S, Polvani G, Bergman O, Eriksson P, Tremoli E. Prenylcysteine oxidase 1, an emerging player in atherosclerosis. Commun Biol 2021; 4:1109. [PMID: 34548610 PMCID: PMC8455616 DOI: 10.1038/s42003-021-02630-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 09/02/2021] [Indexed: 02/08/2023] Open
Abstract
The research into the pathophysiology of atherosclerosis has considerably increased our understanding of the disease complexity, but still many questions remain unanswered, both mechanistically and pharmacologically. Here, we provided evidence that the pro-oxidant enzyme Prenylcysteine Oxidase 1 (PCYOX1), in the human atherosclerotic lesions, is both synthesized locally and transported within the subintimal space by proatherogenic lipoproteins accumulating in the arterial wall during atherogenesis. Further, Pcyox1 deficiency in Apoe-/- mice retards atheroprogression, is associated with decreased features of lesion vulnerability and lower levels of lipid peroxidation, reduces plasma lipid levels and inflammation. PCYOX1 silencing in vitro affects the cellular proteome by influencing multiple functions related to inflammation, oxidative stress, and platelet adhesion. Collectively, these findings identify the pro-oxidant enzyme PCYOX1 as an emerging player in atherogenesis and, therefore, understanding the biology and mechanisms of all functions of this unique enzyme is likely to provide additional therapeutic opportunities in addressing atherosclerosis.
Collapse
Affiliation(s)
- C. Banfi
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - R. Baetta
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - S. S. Barbieri
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - M. Brioschi
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - A. Guarino
- grid.418230.c0000 0004 1760 1750Cardiovascular Tissue Bank of Milan, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - S. Ghilardi
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - L. Sandrini
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - S. Eligini
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - G. Polvani
- grid.418230.c0000 0004 1760 1750Cardiovascular Tissue Bank of Milan, Centro Cardiologico Monzino IRCCS, Milano, Italy ,grid.4708.b0000 0004 1757 2822Department of Clinical Sciences and Community Health, Cardiovascular Section, University of Milan, Milano, Italy ,grid.418230.c0000 0004 1760 1750Department of Cardiovascular Disease, Development and Innovation Cardiac Surgery Unit, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - O. Bergman
- grid.4714.60000 0004 1937 0626Department of Medicine Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - P. Eriksson
- grid.4714.60000 0004 1937 0626Department of Medicine Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - E. Tremoli
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| |
Collapse
|
17
|
Sommariva E, Stadiotti I, Casella M, Catto V, Dello Russo A, Carbucicchio C, Arnaboldi L, De Metrio S, Milano G, Scopece A, Casaburo M, Andreini D, Mushtaq S, Conte E, Chiesa M, Birchmeier W, Cogliati E, Paolin A, König E, Meraviglia V, De Musso M, Volani C, Cattelan G, Rauhe W, Turnu L, Porro B, Pedrazzini M, Camera M, Corsini A, Tondo C, Rossini A, Pompilio G. Oxidized LDL-dependent pathway as new pathogenic trigger in arrhythmogenic cardiomyopathy. EMBO Mol Med 2021; 13:e14365. [PMID: 34337880 PMCID: PMC8422076 DOI: 10.15252/emmm.202114365] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 12/30/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is hallmarked by ventricular fibro-adipogenic alterations, contributing to cardiac dysfunctions and arrhythmias. Although genetically determined (e.g., PKP2 mutations), ACM phenotypes are highly variable. More data on phenotype modulators, clinical prognosticators, and etiological therapies are awaited. We hypothesized that oxidized low-density lipoprotein (oxLDL)-dependent activation of PPARγ, a recognized effector of ACM adipogenesis, contributes to disease pathogenesis. ACM patients showing high plasma concentration of oxLDL display severe clinical phenotypes in terms of fat infiltration, ventricular dysfunction, and major arrhythmic event risk. In ACM patient-derived cardiac cells, we demonstrated that oxLDLs are major cofactors of adipogenesis. Mechanistically, the increased lipid accumulation is mediated by oxLDL cell internalization through CD36, ultimately resulting in PPARγ upregulation. By boosting oxLDL in a Pkp2 heterozygous knock-out mice through high-fat diet feeding, we confirmed in vivo the oxidized lipid dependency of cardiac adipogenesis and right ventricle systolic impairment, which are counteracted by atorvastatin treatment. The modulatory role of oxidized lipids on ACM adipogenesis, demonstrated at cellular, mouse, and patient levels, represents a novel risk stratification tool and a target for ACM pharmacological strategies.
Collapse
Affiliation(s)
- Elena Sommariva
- Unit of Vascular Biology and Regenerative MedicineCentro Cardiologico Monzino IRCCSMilanItaly
| | - Ilaria Stadiotti
- Unit of Vascular Biology and Regenerative MedicineCentro Cardiologico Monzino IRCCSMilanItaly
| | - Michela Casella
- Heart Rhythm CenterCentro Cardiologico Monzino IRCCSMilanItaly
| | - Valentina Catto
- Heart Rhythm CenterCentro Cardiologico Monzino IRCCSMilanItaly
| | | | | | - Lorenzo Arnaboldi
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Simona De Metrio
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Giuseppina Milano
- Unit of Vascular Biology and Regenerative MedicineCentro Cardiologico Monzino IRCCSMilanItaly
- Department of Heart and VesselsLaboratory of Cardiovascular ResearchUniversity Hospital of LausanneLausanneSwitzerland
| | - Alessandro Scopece
- Unit of Vascular Biology and Regenerative MedicineCentro Cardiologico Monzino IRCCSMilanItaly
| | - Manuel Casaburo
- Unit of Vascular Biology and Regenerative MedicineCentro Cardiologico Monzino IRCCSMilanItaly
| | - Daniele Andreini
- Unit of Cardiovascular ImagingCentro Cardiologico Monzino IRCCSMilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Saima Mushtaq
- Unit of Cardiovascular ImagingCentro Cardiologico Monzino IRCCSMilanItaly
| | - Edoardo Conte
- Unit of Cardiovascular ImagingCentro Cardiologico Monzino IRCCSMilanItaly
| | - Mattia Chiesa
- Bioinformatics and Artificial Intelligence facilityCentro Cardiologico Monzino IRCCSMilanItaly
| | | | | | | | - Eva König
- Institute for BiomedicineEurac ResearchAffiliated Institute of the University of LübeckBozenItaly
| | - Viviana Meraviglia
- Institute for BiomedicineEurac ResearchAffiliated Institute of the University of LübeckBozenItaly
| | - Monica De Musso
- Institute for BiomedicineEurac ResearchAffiliated Institute of the University of LübeckBozenItaly
| | - Chiara Volani
- Institute for BiomedicineEurac ResearchAffiliated Institute of the University of LübeckBozenItaly
| | - Giada Cattelan
- Institute for BiomedicineEurac ResearchAffiliated Institute of the University of LübeckBozenItaly
| | | | - Linda Turnu
- Unit of Metabolomics and Cellular Biochemistry of AtherothrombosisCentro Cardiologico Monzino IRCCSMilanItaly
| | - Benedetta Porro
- Unit of Metabolomics and Cellular Biochemistry of AtherothrombosisCentro Cardiologico Monzino IRCCSMilanItaly
| | - Matteo Pedrazzini
- Laboratory of Cardiovascular GeneticsIstituto Auxologico ItalianoIRCCSMilanItaly
| | - Marina Camera
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
- Unit of Cell and Molecular Biology in Cardiovascular DiseasesCentro Cardiologico Monzino IRCCSMilanItaly
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
- IRCCS MultiMedicaMilanItaly
| | - Claudio Tondo
- Heart Rhythm CenterCentro Cardiologico Monzino IRCCSMilanItaly
- Department of BiomedicalSurgical and Dental SciencesUniversità degli Studi di MilanoMilanItaly
| | - Alessandra Rossini
- Institute for BiomedicineEurac ResearchAffiliated Institute of the University of LübeckBozenItaly
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative MedicineCentro Cardiologico Monzino IRCCSMilanItaly
- Department of BiomedicalSurgical and Dental SciencesUniversità degli Studi di MilanoMilanItaly
| |
Collapse
|
18
|
Silva PPB, Pereira RMR, Takayama L, Borba CG, Duarte FH, Trarbach EB, Martin RM, Bronstein MD, Tritos NA, Jallad RS. Impaired Bone Microarchitecture in Premenopausal Women With Acromegaly: The Possible Role of Wnt Signaling. J Clin Endocrinol Metab 2021; 106:2690-2706. [PMID: 33871626 DOI: 10.1210/clinem/dgab260] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Indexed: 12/17/2022]
Abstract
CONTEXT Acromegaly can impair bone integrity, increasing the risk of vertebral fractures (VFs). OBJECTIVE To evaluate the impact of isolated GH/IGF-I hypersecretion on bone turnover markers, Wnt inhibitors, bone mineral density (BMD), microarchitecture, bone strength and vertebral fractures in female patients with acromegaly (Acro), compared with healthy control group (HC). DESIGN, SETTING, AND PATIENTS Cross-sectional study including 83 premenopausal women without any pituitary deficiency:18 acromegaly in remission (AcroR), 12 in group with active acromegaly (AcroA), and 53 HC. Serum procollagen type 1 N-terminal propeptide, β-carboxy-terminal crosslinked telopeptide of type 1 collagen, osteocalcin, sclerostin, and DKK1 were measured in blood samples. dual-energy X-ray absorptiometry, high-resolution peripheral quantitative computed tomography (HR-pQCT) and vertebral fractures evaluation were also assessed simultaneously. MAIN OUTCOME AND RESULTS AcroA showed significantly lower sclerostin and higher DKK1 compared with HC. On HR-pQCT of tibia and radius, Acro showed impairment of trabecular (area and trabecular number), increased cortical porosity, and increased cortical area and cortical thickness compared with HC. The only significant correlation found with HR-pQCT parameters was a positive correlation between cortical porosity and serum DKK1 (R = 0.45, P = 0.044). Mild VFs were present in approximately 30% of patients. CONCLUSIONS Eugonadal women with acromegaly without any pituitary deficiency showed increased cortical BMD, impairment of trabecular bone microstructure, and increased VF. Sclerostin was not correlated with any HR-pQCT parameters; however, DKK1 was correlated with cortical porosity in tibia (P = 0.027). Additional studies are needed to clarify the role of Wnt inhibitors on bone microarchitecture impairment in acromegaly.
Collapse
Affiliation(s)
- Paula P B Silva
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, SP 05403-010, Brazil
| | - Rosa M R Pereira
- Bone Metabolism Laboratory, Rheumatology Division, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, SP 01246903, Brazil
- Rheumatology Division Hospital das Clinicas HCFMUSP, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, SP 05403-010, Brazil
| | - Liliam Takayama
- Rheumatology Division Hospital das Clinicas HCFMUSP, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, SP 05403-010, Brazil
| | - Clarissa G Borba
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, SP 05403-010, Brazil
| | - Felipe H Duarte
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, SP 05403-010, Brazil
| | - Ericka B Trarbach
- Laboratorio de Endocrinologia Celular e Molecular/LIM25, Disciplina de Endocrinologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP 01246903, Brasil
| | - Regina Matsunaga Martin
- Diseases Unit Osteometabolic, Endocrinology Service, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC-FMUSP), São Paulo, SP 05403-010, Brazil
| | - Marcello D Bronstein
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, SP 05403-010, Brazil
- Laboratorio de Endocrinologia Celular e Molecular/LIM25, Disciplina de Endocrinologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP 01246903, Brasil
| | - Nicholas A Tritos
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Raquel S Jallad
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, SP 05403-010, Brazil
- Laboratorio de Endocrinologia Celular e Molecular/LIM25, Disciplina de Endocrinologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP 01246903, Brasil
| |
Collapse
|
19
|
CD276 is an important player in macrophage recruitment into the tumor and an upstream regulator for PAI-1. Sci Rep 2021; 11:14849. [PMID: 34290311 PMCID: PMC8295264 DOI: 10.1038/s41598-021-94360-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
More than 70% of colorectal, prostate, ovarian, pancreatic and breast cancer specimens show expression of CD276 (B7–H3), a potential immune checkpoint family member. Several studies have shown that high CD276 expression in cancer cells correlates with a poor clinical prognosis. This has been associated with the presence of lower tumor infiltrating leukocytes. Among those, tumor-associated macrophages can comprise up to 50% of the tumor mass and are thought to support tumor growth through various mechanisms. However, a lack of information on CD276 function and interaction partner(s) impedes rigorous evaluation of CD276 as a therapeutic target in oncology. Therefore, we aimed to understand the relevance of CD276 in tumor-macrophage interaction by employing a 3D spheroid coculture system with human cells. Our data show a role for tumor-expressed CD276 on the macrophage recruitment into the tumor spheroid, and also in regulation of the extracellular matrix modulator PAI-1. Furthermore, our experiments focusing on macrophage-expressed CD276 suggest that the antibody-dependent CD276 engagement triggers predominantly inhibitory signaling networks in human macrophages.
Collapse
|
20
|
Effect of Statins on Platelet Activation and Function: From Molecular Pathways to Clinical Effects. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6661847. [PMID: 33564680 PMCID: PMC7850835 DOI: 10.1155/2021/6661847] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
Purpose Statins are a class of drugs widely used in clinical practice for their lipid-lowering and pleiotropic effects. In recent years, a correlation between statins and platelet function has been unveiled in the literature that might introduce new therapeutic indications for this class of drugs. This review is aimed at summarizing the mechanisms underlying statin-platelet interaction in the cardiologic scenario and building the basis for future in-depth studies. Methods We conducted a literature search through PubMed, Embase, EBSCO, Cochrane Database of Systematic Reviews, and Web of Science from their inception to June 2020. Results Many pathways could explain the interaction between statins and platelets, but the specific effect depends on the specific compound. Some could be mediated by enzymes that allow the entry of drugs into the cell (OATP2B1) and others by enzymes that mediate their activation (PLA2, MAPK, TAX2, PPARs, AKT, and COX-1), recruitment and adhesion (LOX-1, CD36, and CD40L), or apoptosis (BCL2). Statins also appear to have a synergistic effect with aspirin and low molecular weight heparins. Surprisingly, they seem to have an antagonistic effect with clopidogrel. Conclusion There are many pathways potentially responsible for the interactions between statins and platelets. Their effect appears to be closely related, and each single effect can be barely measured. Also, the same compound might have complex downstream signaling with potentially opposite effects, i.e., beneficial or deleterious. The multiple clinical implications that can be derived as a result of this interaction, however, represent an excellent reason to develop future in-depth studies.
Collapse
|
21
|
Cattaneo MG, Banfi C, Brioschi M, Lattuada D, Vicentini LM. Sex-dependent differences in the secretome of human endothelial cells. Biol Sex Differ 2021; 12:7. [PMID: 33413676 PMCID: PMC7791663 DOI: 10.1186/s13293-020-00350-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cellular sex has rarely been considered as a biological variable in preclinical research, even when the pathogenesis of diseases with predictable sex differences is studied. In this perspective, proteomics, and “omics” approaches in general, can provide powerful tools to obtain comprehensive cellular maps, thus favoring the discovery of still unknown sex-biased physio-pathological mechanisms. Methods We performed proteomic and Gene Ontology (GO) analyses of the secretome from human serum-deprived male and female endothelial cells (ECs) followed by ELISA validation. Apoptosis was detected by FACS and Western blot techniques and efferocytosis through the ability of the macrophage cell line RAW 264.7 to engulf apoptotic ECs. PTX3 mRNA levels were measured by RT-qPCR. Results Proteomic and GO analyses of the secretome from starved human male and female ECs demonstrated a significant enrichment in proteins related to cellular responses to stress and to the regulation of apoptosis in the secretome of male ECs. Accordingly, a higher percentage of male ECs underwent apoptosis in response to serum deprivation in comparison with female ECs. Among the secreted proteins, we reliably found higher levels of PTX3 in the male EC secretome. The silencing of PTX3 suggested that male ECs were dependent on its expression to properly carry out the efferocytotic process. At variance, female EC efferocytosis seemed to be independent on PTX3 expression. Conclusions Our results demonstrated that serum-starved male and female ECs possess different secretory phenotypes that might take part in the sex-biased response to cellular stress. We identified PTX3 as a crucial player in the male-specific endothelial response to an apoptotic trigger. This novel and sex-related role for secreted proteins, and mainly for PTX3, may open the way to the discovery of still unknown sex-specific mechanisms and pharmacological targets for the prevention and treatment of endothelial dysfunction at the onset of atherosclerosis and cardiovascular disease. Supplementary Information The online version contains supplementary material available at 10.1186/s13293-020-00350-3.
Collapse
Affiliation(s)
- Maria Grazia Cattaneo
- Dept of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Vanvitelli 32, Milan, Italy.
| | | | | | - Donatella Lattuada
- Dept of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Vanvitelli 32, Milan, Italy
| | - Lucia M Vicentini
- Dept of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Vanvitelli 32, Milan, Italy
| |
Collapse
|
22
|
Niedzielski M, Broncel M, Gorzelak-Pabiś P, Woźniak E. New possible pharmacological targets for statins and ezetimibe. Biomed Pharmacother 2020; 129:110388. [PMID: 32559626 DOI: 10.1016/j.biopha.2020.110388] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/30/2020] [Accepted: 06/07/2020] [Indexed: 12/25/2022] Open
Abstract
Statin therapy is the gold standard in the treatment of dyslipidemia. Understanding the mechanisms of action of these drugs provides an opportunity to define new therapeutic goals for pharmacotherapy in patients with atherosclerotic lesions. The present review indicates the existence of previously unknown therapeutic targets for statins, such as Krüppel-like Factor 2 (KLF-2), Cystathionine γ lyase (CSE) and the microRNA regulating eNOS activity and synthesis; nuclear PXR receptor and EB transcription factor regulating Inflammasome NLRP3 activity; the Dickkopf-related protein 1 (DKK-1), which inhibits the WNT signalling pathway; the peroxisome proliferator-activated receptor (PPAR-γ) in vascular smooth muscle cells (VSMCs), which regulates the cell cycle, and the ERK5-Nrf2 pathway, which reduces the level of harmful advanced glycation end-products (AGE) in VSMCs during diabetic vasculopathy. Importantly, our review includes a number of promising discoveries, specifically those related to the effects of miR-221, miR-222 and miR-27b on the structure, synthesis and activity of eNOS, such as microRNA-based therapies, which offer promise in future targeted therapies. In contrast to numerous experiments confirming the pleiotropic effect of statins, there is still insufficient evidence on the pleiotropic effect of ezetimibe, which goes beyond its basic inhibitory effect on intestinal cholesterol absorption. However, recent studies indicate that this effect is limited to inhibiting macrophage migration, decreasing VCAM-1 expression and reducing the levels of reactive oxygen species. Defining new therapeutic goals for pharmacotherapy in patients with atherosclerotic lesions and ensuring effective treatment of dyslipidemia and its associated cardiovascular complications requires a thorough understanding of both the mechanisms of action of these drugs and of atherosclerosis itself.
Collapse
Affiliation(s)
- Mateusz Niedzielski
- Medical University of Lodz, Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Kniaziewicza 1/5, 91-347 Lodz, Poland
| | - Marlena Broncel
- Medical University of Lodz, Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Kniaziewicza 1/5, 91-347 Lodz, Poland
| | - Paulina Gorzelak-Pabiś
- Medical University of Lodz, Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Kniaziewicza 1/5, 91-347 Lodz, Poland
| | - Ewelina Woźniak
- Medical University of Lodz, Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Kniaziewicza 1/5, 91-347 Lodz, Poland.
| |
Collapse
|
23
|
Boucher P, Matz RL, Terrand J. atherosclerosis: gone with the Wnt? Atherosclerosis 2020; 301:15-22. [PMID: 32289618 DOI: 10.1016/j.atherosclerosis.2020.03.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/19/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022]
Abstract
Atherosclerosis, a pathology affecting large and medium-sized arteries, is the major cause of cardiovascular morbidity/mortality in industrialized countries. During atherosclerosis, cells accumulate large amounts of cholesterol through the uptake of modified low-density lipoprotein particles to form foam cells. This accumulation forms the basis for the development of the disease and for a large spectrum of other diseases in various organs. Massive research efforts have yielded valuable information about the underlying molecular mechanisms of atherosclerosis. In particular, newer discoveries on the early stage of lesion formation, cholesterol accumulation, reverse cholesterol transport, and local inflammation in the vascular wall have opened unanticipated horizons of understanding and raised novel questions and therapeutic opportunities. In this review, we focus on Wnt signaling, which has received little attention so far, yet affects lysosomal function and signalling pathways that limit cholesterol accumulation. This occurs in different tissues and cell types, including smooth muscle cells, endothelial cells and macrophages in the arterial wall, and thus profoundly impacts on atherosclerotic disease development and progression.
Collapse
Affiliation(s)
- Philippe Boucher
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France.
| | - Rachel L Matz
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France
| | - Jérôme Terrand
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France
| |
Collapse
|
24
|
Klingenschmid G, Tschiderer L, Himmler G, Rungger G, Brugger S, Santer P, Willeit J, Kiechl S, Willeit P. Associations of Serum Dickkopf-1 and Sclerostin With Cardiovascular Events: Results From the Prospective Bruneck Study. J Am Heart Assoc 2020; 9:e014816. [PMID: 32172649 PMCID: PMC7335516 DOI: 10.1161/jaha.119.014816] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Dickkopf-1 and sclerostin have been implicated in atherosclerosis and vascular calcification. We aimed to quantify the association of their serum levels with incident cardiovascular disease (CVD) in the general population. Methods and Results Among 706 participants of the prospective, population-based Bruneck Study, mean±SD of serum levels were 44.5±14.7 pmol/L for dickkopf-1 and 47.1±17.5 pmol/L for sclerostin. The primary outcome was a composite CVD end point composed of ischemic or hemorrhagic stroke, transient ischemic attack, myocardial infarction, angina pectoris, peripheral vascular disease, and revascularization procedures. Over a median follow-up duration of 15.6 years, 179 CVD events occurred. For the primary CVD outcome, multivariable-adjusted hazard ratios (HRs) per SD higher level were 1.20 for dickkopf-1 (95% CI, 1.02-1.42; P=0.028) and 0.92 for sclerostin (95% CI, 0.78-1.08; P=0.286). Secondary outcome analyses revealed that the association of dickkopf-1 was primarily driven by ischemic and hemorrhagic stroke (67 events; HR, 1.37; 95% CI, 1.06-1.78; P=0.017), whereas no increase in risk was observed for transient ischemic attack (22 events; HR, 0.87; 95% CI, 0.53-1.44; P=0.593), myocardial infarction (45 events; HR, 1.10; 95% CI, 0.78-1.54; P=0.598), or for other CVD (45 events; HR, 1.25; 95% CI, 0.88-1.76; P=0.209). Conclusions In this prospective, population-based study, elevated baseline levels of dickkopf-1, but not sclerostin, were independently associated with incident cardiovascular events, which was mainly driven by stroke. Our findings support the hypothesis of a role of dickkopf-1 in the pathogenesis of CVD.
Collapse
Affiliation(s)
| | - Lena Tschiderer
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | | | | | - Stefan Brugger
- Departments of Internal Medicine Hospital of Bruneck Italy
| | - Peter Santer
- Departments of Laboratory Medicine Hospital of Bruneck Italy
| | - Johann Willeit
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Stefan Kiechl
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Peter Willeit
- Department of Neurology Medical University of Innsbruck Innsbruck Austria.,Department of Public Health and Primary Care University of Cambridge United Kingdom
| |
Collapse
|
25
|
Martinez Fernandez A, Regazzoni L, Brioschi M, Gianazza E, Agostoni P, Aldini G, Banfi C. Pro-oxidant and pro-inflammatory effects of glycated albumin on cardiomyocytes. Free Radic Biol Med 2019; 144:245-255. [PMID: 31260731 DOI: 10.1016/j.freeradbiomed.2019.06.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 06/13/2019] [Accepted: 06/19/2019] [Indexed: 12/14/2022]
Abstract
Human serum albumin (HSA) is the most abundant circulating protein in the body and presents an extensive range of biological functions. As such, it is prone to undergo post-translational modifications (PTMs). The non-enzymatic early glycation of HSA, one of the several PTMs undergone by HSA, arises from the addition of reducing sugars to amine group residues, thus modifying the structure of HSA. These changes may affect HSA functions impairing its biological activity, finally leading to cell damage. The aim of this study was to quantitate glycated-HSA (GA) levels in the plasma of heart failure (HF) patients and to evaluate the biological effects of GA on HL-1 cardiomyocytes. Plasma GA content from HF patients and healthy subjects was measured by direct infusion electrospray ionization mass spectrometry (ESI-MS). Results pointed out a significant increase of GA in HF patients with respect to the control group (p < 0.05). Additionally, after stimulation with GA, proteomic analysis of HL-1 secreted proteins showed the modulation of several proteins involved, among other processes, in the response to stress. Further, stimulated cells showed a rapid increase in ROS generation, higher mRNA levels of the inflammatory cytokine interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α), and higher levels of the oxidative 4-HNE-protein adducts and carbonylated proteins. Our findings show that plasma GA is increased in HF patients. Further, GA exerts pro-inflammatory and pro-oxidant effects on cardiomyocytes, which suggest a causal role in the etiopathogenesis of HF.
Collapse
MESH Headings
- Aged
- Case-Control Studies
- Cell Death
- Cell Line
- Dyslipidemias/blood
- Dyslipidemias/genetics
- Dyslipidemias/pathology
- Female
- Gene Expression Profiling
- Gene Ontology
- Glycation End Products, Advanced
- Glycosylation
- HSP90 Heat-Shock Proteins/genetics
- HSP90 Heat-Shock Proteins/metabolism
- Heart Failure/blood
- Heart Failure/genetics
- Heart Failure/pathology
- Humans
- Hypertension/blood
- Hypertension/genetics
- Hypertension/pathology
- Interleukin-6/genetics
- Interleukin-6/metabolism
- Lysine/analogs & derivatives
- Lysine/blood
- Male
- Middle Aged
- Molecular Sequence Annotation
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Natriuretic Peptide, Brain/blood
- Natriuretic Peptide, Brain/genetics
- Protein Carbonylation
- Protein Processing, Post-Translational
- Reactive Oxygen Species/agonists
- Reactive Oxygen Species/metabolism
- Serum Albumin/pharmacology
- Serum Albumin, Human/chemistry
- Serum Albumin, Human/genetics
- Serum Albumin, Human/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Glycated Serum Albumin
Collapse
Affiliation(s)
| | - Luca Regazzoni
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | | | | | - Piergiuseppe Agostoni
- Centro Cardiologico Monzino, IRCCS, Milan, Italy; Department of Clinical Sciences and Community Health, Cardiovascular Section, University of Milan, Milan, Italy
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | | |
Collapse
|
26
|
Palmer MA, Blakeborough L, Harries M, Haslam IS. Cholesterol homeostasis: Links to hair follicle biology and hair disorders. Exp Dermatol 2019; 29:299-311. [PMID: 31260136 DOI: 10.1111/exd.13993] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/24/2019] [Accepted: 06/19/2019] [Indexed: 01/10/2023]
Abstract
Lipids and lipid metabolism are critical factors in hair follicle (HF) biology, and cholesterol has long been suspected of influencing hair growth. Altered cholesterol homeostasis is involved in the pathogenesis of primary cicatricial alopecia, mutations in a cholesterol transporter are associated with congenital hypertrichosis, and dyslipidaemia has been linked to androgenic alopecia. The underlying molecular mechanisms by which cholesterol influences pathways involved in proliferation and differentiation within HF cell populations remain largely unknown. As such, expanding our knowledge of the role for cholesterol in regulating these processes is likely to provide new leads in the development of treatments for disorders of hair growth and cycling. This review describes the current state of knowledge with respect to cholesterol homeostasis in the HF along with known and putative links to hair pathologies.
Collapse
Affiliation(s)
- Megan A Palmer
- School of Applied Sciences, Department of Biological and Geographical Sciences, University of Huddersfield, Huddersfield, UK
| | - Liam Blakeborough
- School of Applied Sciences, Department of Biological and Geographical Sciences, University of Huddersfield, Huddersfield, UK
| | - Matthew Harries
- Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.,Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Iain S Haslam
- School of Applied Sciences, Department of Biological and Geographical Sciences, University of Huddersfield, Huddersfield, UK
| |
Collapse
|
27
|
Li Y, Meng R. MicroRNA-154 Targets the Wnt/β-Catenin Signaling Pathway Following Injury to Human Vascular Endothelial Cells by Hydrogen Peroxide. Med Sci Monit 2019; 25:5648-5656. [PMID: 31359876 PMCID: PMC6685327 DOI: 10.12659/msm.915263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Endothelial cells are involved in vascular homeostasis, and endothelial cell dysfunction is involved in the pathogenesis of cardiovascular disease. This study aimed to investigate the effects of microRNA-154 in human umbilical vein endothelial cells (HUVECs) following injury induced by hydrogen peroxide (H2O2). Material/Methods Cell viability and apoptosis of HUVECs treated with H2O2 were measured. The expression of microRNA-154 was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Cell survival, caspase-3 activity, and the apoptosis rate were evaluated in H2O2-treated HUVECs cells after the upregulation and down-regulation of microRNA-154 expression. The interaction between microRNA-154 and Dickkopf WNT signaling pathway inhibitor 2 (DKK2) was predicted by bioinformatics analysis and was verified by luciferase reporter gene assay and Western blot. The effects of DKK2 short-interfering RNA (siRNA) on antioxidant injury in HUVECs cells were determined. Results The survival rate of HUVECs exposed to H2O2 was significantly reduced and the apoptosis rate was significantly increased, and H2O2 significantly inhibited the expression of microRNA-154 in a dose-dependent manner. Overexpression of microRNA-154 increased cell survival, reduced the activity of caspase-3, and reduced cell apoptosis. Inhibition of microRNA-154 expression decreased cell survival, increased the activity of caspase-3, and promoted cell apoptosis. Luciferase reporter gene assay and Western blot showed that microRNA-154 interacted with the Wnt pathway molecule DKK2 in HUVECS. Also, DDK2 siRNA resulted in a similar protective effect on H2O2-treated HUVECs as overexpression of microRNA-154. Conclusions Oxidative injury in HUVECs was regulated by microRNA-154 targeting the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yan Li
- Department of Cardiology, The First Peoples' Hospital of Jining, Jining, Shandong, China (mainland)
| | - Ranran Meng
- Department of Vascular Surgery, The First Peoples' Hospital of Jining, Jining, Shandong, China (mainland)
| |
Collapse
|
28
|
Abstract
Clinical and preclinical studies over the past 3 decades have uncovered a multitude of signaling pathways involved in the initiation and progression of atherosclerosis. From these studies, signaling by proteins of the Wnt family has recently emerged as an important player in the development of atherosclerosis. Wnt signaling is characterized by a large number of ligands, receptors, and coreceptors and can be regulated at many different levels. Among Wnt modulators, the evolutionary conserved Dkk (Dickkopf) proteins, and especially Dkk-1, the founding member of the family, are the best characterized. The role of Dkks in the pathophysiology of the arterial wall is only partially understood, but their involvement in atherosclerosis is becoming increasingly evident. This review introduces recent key findings on Dkk proteins and their functions in atherosclerosis and discusses the potential importance of modulating Dkk signaling as part of a novel, improved strategy for preventing and treating atherosclerosis-related diseases.
Visual Overview—
An online visual overview is available for this article.
Collapse
Affiliation(s)
- Roberta Baetta
- From the Centro Cardiologico Monzino, IRCCS, Milano, Italy
| | - Cristina Banfi
- From the Centro Cardiologico Monzino, IRCCS, Milano, Italy
| |
Collapse
|