1
|
van der Linden J, Stefens SJM, Heredia‐Genestar JM, Ridwan Y, Brandt RMC, van Vliet N, de Beer I, van Thiel BS, Steen H, Cheng C, Roks AJM, Danser AHJ, Essers J, van der Pluijm I. Ercc1 DNA repair deficiency results in vascular aging characterized by VSMC phenotype switching, ECM remodeling, and an increased stress response. Aging Cell 2024; 23:e14126. [PMID: 38451018 PMCID: PMC11113264 DOI: 10.1111/acel.14126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Cardiovascular diseases are the number one cause of death globally. The most important determinant of cardiovascular health is a person's age. Aging results in structural changes and functional decline of the cardiovascular system. DNA damage is an important contributor to the aging process, and mice with a DNA repair defect caused by Ercc1 deficiency display hypertension, vascular stiffening, and loss of vasomotor control. To determine the underlying cause, we compared important hallmarks of vascular aging in aortas of both Ercc1Δ/- and age-matched wildtype mice. Additionally, we investigated vascular aging in 104 week old wildtype mice. Ercc1Δ/- aortas displayed arterial thickening, a loss of cells, and a discontinuous endothelial layer. Aortas of 24 week old Ercc1Δ/- mice showed phenotypical switching of vascular smooth muscle cells (VSMCs), characterized by a decrease in contractile markers and a decrease in synthetic markers at the RNA level. As well as an increase in osteogenic markers, microcalcification, and an increase in markers for damage induced stress response. This suggests that Ercc1Δ/- VSMCs undergo a stress-induced contractile-to-osteogenic phenotype switch. Ercc1Δ/- aortas showed increased MMP activity, elastin fragmentation, and proteoglycan deposition, characteristic of vascular aging and indicative of age-related extracellular matrix remodeling. The 104 week old WT mice showed loss of cells, VSMC dedifferentiation, and senescence. In conclusion, Ercc1Δ/- aortas rapidly display many characteristics of vascular aging, and thus the Ercc1Δ/- mouse is an excellent model to evaluate drugs that prevent vascular aging in a short time span at the functional, histological, and cellular level.
Collapse
Affiliation(s)
- Janette van der Linden
- Division of Vascular Medicine and Pharmacology, Department of Internal MedicineErasmus University Medical CenterRotterdamThe Netherlands
- Department of Molecular Genetics, Cancer Genomics CenterErasmus University Medical CenterRotterdamThe Netherlands
| | - Sanne J. M. Stefens
- Department of Molecular Genetics, Cancer Genomics CenterErasmus University Medical CenterRotterdamThe Netherlands
| | - José María Heredia‐Genestar
- Department of Molecular Genetics, Cancer Genomics CenterErasmus University Medical CenterRotterdamThe Netherlands
| | - Yanto Ridwan
- Department of Molecular Genetics, Cancer Genomics CenterErasmus University Medical CenterRotterdamThe Netherlands
- AMIE Core facilityErasmus University Medical CenterRotterdamThe Netherlands
| | - Renata M. C. Brandt
- Department of Molecular Genetics, Cancer Genomics CenterErasmus University Medical CenterRotterdamThe Netherlands
| | - Nicole van Vliet
- Department of Molecular Genetics, Cancer Genomics CenterErasmus University Medical CenterRotterdamThe Netherlands
| | - Isa de Beer
- Department of Molecular Genetics, Cancer Genomics CenterErasmus University Medical CenterRotterdamThe Netherlands
| | - Bibi S. van Thiel
- Department of Molecular Genetics, Cancer Genomics CenterErasmus University Medical CenterRotterdamThe Netherlands
| | | | - Caroline Cheng
- Division of Experimental Cardiology, Department of CardiologyMC UtrechtUtrechtThe Netherlands
- Division of Internal Medicine and Dermatology, Department of Nephrology and HypertensionMC UtrechtUtrechtThe Netherlands
| | - Anton J. M. Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal MedicineErasmus University Medical CenterRotterdamThe Netherlands
| | - A. H. Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal MedicineErasmus University Medical CenterRotterdamThe Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Cancer Genomics CenterErasmus University Medical CenterRotterdamThe Netherlands
- Department of Vascular SurgeryCardiovascular Institute, Erasmus University Medical CenterRotterdamThe Netherlands
- Department of RadiotherapyErasmus University Medical CenterRotterdamThe Netherlands
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Cancer Genomics CenterErasmus University Medical CenterRotterdamThe Netherlands
- Department of Vascular SurgeryCardiovascular Institute, Erasmus University Medical CenterRotterdamThe Netherlands
| |
Collapse
|
2
|
Evenepoel P, Stenvinkel P, Shanahan C, Pacifici R. Inflammation and gut dysbiosis as drivers of CKD-MBD. Nat Rev Nephrol 2023; 19:646-657. [PMID: 37488276 DOI: 10.1038/s41581-023-00736-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
Two decades ago, Kidney Disease: Improving Global Outcomes coined the term chronic kidney disease-mineral and bone disorder (CKD-MBD) to describe the syndrome of biochemical, bone and extra-skeletal calcification abnormalities that occur in patients with CKD. CKD-MBD is a prevalent complication and contributes to the excessively high burden of fractures and cardiovascular disease, loss of quality of life and premature mortality in patients with CKD. Thus far, therapy has focused primarily on phosphate retention, abnormal vitamin D metabolism and parathyroid hormone disturbances, but these strategies have largely proved unsuccessful, thus calling for paradigm-shifting concepts and innovative therapeutic approaches. Interorgan crosstalk is increasingly acknowledged to have an important role in health and disease. Accordingly, mounting evidence suggests a role for both the immune system and the gut microbiome in bone and vascular biology. Gut dysbiosis, compromised gut epithelial barrier and immune cell dysfunction are prominent features of the uraemic milieu. These alterations might contribute to the inflammatory state observed in CKD and could have a central role in the pathogenesis of CKD-MBD. The emerging fields of osteoimmunology and osteomicrobiology add another level of complexity to the pathogenesis of CKD-MBD, but also create novel therapeutic opportunities.
Collapse
Affiliation(s)
- Pieter Evenepoel
- Laboratory of Nephrology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Herestraat, Leuven, Belgium.
| | - Peter Stenvinkel
- Department of Renal Medicine M99, Karolinska University Hospital, Stockholm, Sweden
| | - Catherine Shanahan
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory Microbiome Research Center, and Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, GA, USA
| |
Collapse
|
3
|
Zhu Y, Zhang JL, Yan XJ, Ji Y, Wang FF. Exploring a new mechanism between lactate and VSMC calcification: PARP1/POLG/UCP2 signaling pathway and imbalance of mitochondrial homeostasis. Cell Death Dis 2023; 14:598. [PMID: 37679327 PMCID: PMC10484939 DOI: 10.1038/s41419-023-06113-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/17/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023]
Abstract
Lactate leads to the imbalance of mitochondria homeostasis, which then promotes vascular calcification. PARP1 can upregulate osteogenic genes and accelerate vascular calcification. However, the relationship among lactate, PARP1, and mitochondrial homeostasis is unclear. The present study aimed to explore the new molecular mechanism of lactate to promote VSMC calcification by evaluating PARP1 as a breakthrough molecule. A coculture model of VECs and VSMCs was established, and the model revealed that the glycolysis ability and lactate production of VECs were significantly enhanced after incubation in DOM. Osteogenic marker expression, calcium deposition, and apoptosis in VSMCs were decreased after lactate dehydrogenase A knockdown in VECs. Mechanistically, exogenous lactate increased the overall level of PARP and PARylation in VSMCs. PARP1 knockdown inhibited Drp1-mediated mitochondrial fission and partially restored PINK1/Parkin-mediated mitophagy, thereby reducing mitochondrial oxidative stress. Moreover, lactate induced the translocation of PARP1 from the nucleus to the mitochondria, which then combined with POLG and inhibited POLG-mediated mitochondrial DNA synthesis. This process led to the downregulation of mitochondria-encoded genes, disturbance of mitochondrial respiration, and inhibition of oxidative phosphorylation. The knockdown of PARP1 could partially reverse the damage of mitochondrial gene expression and function caused by lactate. Furthermore, UCP2 was upregulated by the PARP1/POLG signal, and UCP2 knockdown inhibited Drp1-mediated mitochondrial fission and partially recovered PINK1/Parkin-mediated mitophagy. Finally, UCP2 knockdown in VSMCs alleviated DOM-caused VSMC calcification in the coculture model. The study results thus suggest that upregulated PARP1 is involved in the mechanism through which lactate accelerates VSMC calcification partly via POLG/UCP2-caused unbalanced mitochondrial homeostasis.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China
| | - Jia-Li Zhang
- Department of Gastroenterology Centre, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, PR China
| | - Xue-Jiao Yan
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China
| | - Yuan Ji
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China.
| | - Fang-Fang Wang
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China.
| |
Collapse
|
4
|
Gusev E, Sarapultsev A. Atherosclerosis and Inflammation: Insights from the Theory of General Pathological Processes. Int J Mol Sci 2023; 24:ijms24097910. [PMID: 37175617 PMCID: PMC10178362 DOI: 10.3390/ijms24097910] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Recent advances have greatly improved our understanding of the molecular mechanisms behind atherosclerosis pathogenesis. However, there is still a need to systematize this data from a general pathology perspective, particularly with regard to atherogenesis patterns in the context of both canonical and non-classical inflammation types. In this review, we analyze various typical phenomena and outcomes of cellular pro-inflammatory stress in atherosclerosis, as well as the role of endothelial dysfunction in local and systemic manifestations of low-grade inflammation. We also present the features of immune mechanisms in the development of productive inflammation in stable and unstable plaques, along with their similarities and differences compared to canonical inflammation. There are numerous factors that act as inducers of the inflammatory process in atherosclerosis, including vascular endothelium aging, metabolic dysfunctions, autoimmune, and in some cases, infectious damage factors. Life-critical complications of atherosclerosis, such as cardiogenic shock and severe strokes, are associated with the development of acute systemic hyperinflammation. Additionally, critical atherosclerotic ischemia of the lower extremities induces paracoagulation and the development of chronic systemic inflammation. Conversely, sepsis, other critical conditions, and severe systemic chronic diseases contribute to atherogenesis. In summary, atherosclerosis can be characterized as an independent form of inflammation, sharing similarities but also having fundamental differences from low-grade inflammation and various variants of canonical inflammation (classic vasculitis).
Collapse
Affiliation(s)
- Evgenii Gusev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080 Chelyabinsk, Russia
| |
Collapse
|
5
|
Sutton NR, Malhotra R, Hilaire C, Aikawa E, Blumenthal RS, Gackenbach G, Goyal P, Johnson A, Nigwekar SU, Shanahan CM, Towler DA, Wolford BN, Chen Y. Molecular Mechanisms of Vascular Health: Insights From Vascular Aging and Calcification. Arterioscler Thromb Vasc Biol 2023; 43:15-29. [PMID: 36412195 PMCID: PMC9793888 DOI: 10.1161/atvbaha.122.317332] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022]
Abstract
Cardiovascular disease is the most common cause of death worldwide, especially beyond the age of 65 years, with the vast majority of morbidity and mortality due to myocardial infarction and stroke. Vascular pathology stems from a combination of genetic risk, environmental factors, and the biologic changes associated with aging. The pathogenesis underlying the development of vascular aging, and vascular calcification with aging, in particular, is still not fully understood. Accumulating data suggests that genetic risk, likely compounded by epigenetic modifications, environmental factors, including diabetes and chronic kidney disease, and the plasticity of vascular smooth muscle cells to acquire an osteogenic phenotype are major determinants of age-associated vascular calcification. Understanding the molecular mechanisms underlying genetic and modifiable risk factors in regulating age-associated vascular pathology may inspire strategies to promote healthy vascular aging. This article summarizes current knowledge of concepts and mechanisms of age-associated vascular disease, with an emphasis on vascular calcification.
Collapse
Affiliation(s)
- Nadia R. Sutton
- Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Rajeev Malhotra
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Cynthia Hilaire
- Division of Cardiology, Departments of Medicine and Bioengineering, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, 1744 BSTWR, 200 Lothrop St, Pittsburgh, PA, 15260 USA
| | - Elena Aikawa
- Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Roger S. Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease; Baltimore, MD
| | - Grace Gackenbach
- Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Parag Goyal
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Adam Johnson
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Sagar U. Nigwekar
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Catherine M. Shanahan
- School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, UK
| | - Dwight A. Towler
- Department of Medicine | Endocrine Division and Pak Center for Mineral Metabolism Research, UT Southwestern Medical Center, Dallas, TX USA
| | - Brooke N. Wolford
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham and Research Department, Veterans Affairs Birmingham Medical Center, Birmingham, AL, USA
| |
Collapse
|
6
|
Collins MT, Marcucci G, Anders HJ, Beltrami G, Cauley JA, Ebeling PR, Kumar R, Linglart A, Sangiorgi L, Towler DA, Weston R, Whyte MP, Brandi ML, Clarke B, Thakker RV. Skeletal and extraskeletal disorders of biomineralization. Nat Rev Endocrinol 2022; 18:473-489. [PMID: 35578027 DOI: 10.1038/s41574-022-00682-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/13/2022] [Indexed: 12/15/2022]
Abstract
The physiological process of biomineralization is complex and deviation from it leads to a variety of diseases. Progress in the past 10 years has enhanced understanding of the genetic, molecular and cellular pathophysiology underlying these disorders; sometimes, this knowledge has both facilitated restoration of health and clarified the very nature of biomineralization as it occurs in humans. In this Review, we consider the principal regulators of mineralization and crystallization, and how dysregulation of these processes can lead to human disease. The knowledge acquired to date and gaps still to be filled are highlighted. The disorders of mineralization discussed comprise a broad spectrum of conditions that encompass bone disorders associated with alterations of mineral quantity and quality, as well as disorders of extraskeletal mineralization (hyperphosphataemic familial tumoural calcinosis). Included are disorders of alkaline phosphatase (hypophosphatasia) and phosphate homeostasis (X-linked hypophosphataemic rickets, fluorosis, rickets and osteomalacia). Furthermore, crystallopathies are covered as well as arterial and renal calcification. This Review discusses the current knowledge of biomineralization derived from basic and clinical research and points to future studies that will lead to new therapeutic approaches for biomineralization disorders.
Collapse
Affiliation(s)
- Michael T Collins
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA.
| | - Gemma Marcucci
- Bone Metabolic Diseases Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Hans-Joachim Anders
- Department of Medicine IV, Hospital of the University of Munich, Ludwig-Maximilians University, Munich, Germany
| | - Giovanni Beltrami
- Department Paediatric Orthopedic Oncology, Careggi and Meyer Children Hospital, Florence, Italy
| | - Jane A Cauley
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia
| | - Rajiv Kumar
- Departments of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Agnès Linglart
- APHP, Endocrinologie et diabète de l'enfant, Paris, France
| | - Luca Sangiorgi
- Medical Genetics and Skeletal Rare Diseases, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Dwight A Towler
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ria Weston
- Cardiovascular Research Group, Manchester Metropolitan University, Manchester, UK
| | - Michael P Whyte
- Center for Metabolic Bone Disease and Molecular Research, Shriners Hospitals for Children-St Louis, St Louis, MO, USA
- Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | | | - Bart Clarke
- Mayo Clinic Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Rochester, MN, USA
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Zhu X, Ma K, Zhou K, Pan X, Liu J, Nürnberg B, Alesutan I, Völkl J, Lang F. Requirement of Na+/H+ Exchanger NHE1 for Vasopressin-Induced Osteogenic Signaling and Calcification in Human Aortic Smooth Muscle Cells. Kidney Blood Press Res 2022; 47:399-409. [PMID: 35339998 DOI: 10.1159/000524050] [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: 11/29/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND/AIMS Vasopressin is a powerful stimulator of vascular calcification, augmenting osteogenic signaling in vascular smooth muscle cells (VSMCs) including upregulation of transcription factors such as core-binding factor α-1 (CBFA1), msh homeobox 2 (MSX2), and SRY-Box 9 (SOX9), as well as of tissue-nonspecific alkaline phosphatase (ALPL). Vasopressin-induced osteogenic signaling and calcification require the serum- and glucocorticoid-inducible kinase 1 (SGK1). Known effects of SGK1 include upregulation of Na+/H+ exchanger 1 (NHE1). NHE1 further participates in the regulation of reactive oxygen species (ROS). NHE1 has been shown to participate in the orchestration of bone mineralization. The present study, thus, explored whether vasopressin modifies NHE1 expression and ROS generation, as well as whether pharmacological inhibition of NHE1 disrupts vasopressin-induced osteogenic signaling and calcification in VSMCs. METHODS Human aortic smooth muscle cells (HAoSMCs) were treated with vasopressin in the absence or presence of SGK1 silencing, SGK1 inhibitor GSK-650394, and NHE1 blocker cariporide. Transcript levels were determined by using quantitative real-time polymerase chain reaction, protein abundance by Western blotting, ROS generation with 2',7'-dichlorofluorescein diacetate fluorescence, and ALP activity and calcium content by using colorimetric assays. RESULTS Vasopressin significantly enhanced the NHE1 transcript and protein levels in HAoSMCs, effects significantly blunted by SGK1 inhibition with GSK-650394 or SGK1 silencing. Vasopressin increased ROS accumulation, an effect significantly blocked by the NHE1 inhibitor cariporide. Vasopressin further significantly increased osteogenic markers CBFA1, MSX2, SOX9, and ALPL transcript levels, as well as ALP activity and calcium content in HAoSMCs, all effects significantly blunted by SGK1 silencing or in the presence of GSK-650394 or cariporide. CONCLUSION Vasopressin stimulates NHE1 expression and ROS generation, an effect dependent on SGK1 and required for vasopressin-induced stimulation of osteogenic signaling and calcification of VSMCs.
Collapse
Affiliation(s)
- Xuexue Zhu
- Department of Pharmacology, Experimental Therapy & Toxicology, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Ke Ma
- Department of Pharmacology, Experimental Therapy & Toxicology, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Kuo Zhou
- Department of Pharmacology, Experimental Therapy & Toxicology, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Xia Pan
- Department of Pharmacology, Experimental Therapy & Toxicology, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Jibin Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bernd Nürnberg
- Department of Pharmacology, Experimental Therapy & Toxicology, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Jakob Völkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria.,Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Florian Lang
- Department of Physiology, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| |
Collapse
|
8
|
Chen C, Wu Y, Lu HL, Liu K, Qin X. Identification of potential biomarkers of vascular calcification using bioinformatics analysis and validation in vivo. PeerJ 2022; 10:e13138. [PMID: 35313524 PMCID: PMC8934046 DOI: 10.7717/peerj.13138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/28/2022] [Indexed: 01/12/2023] Open
Abstract
Background Vascular calcification (VC) is the most widespread pathological change in diseases of the vascular system. However, we know poorly about the molecular mechanisms and effective therapeutic approaches of VC. Methods The VC dataset, GSE146638, was downloaded from the Gene Expression Omnibus (GEO) database. Using the edgeR package to screen Differentially expressed genes (DEGs). Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) were used to find pathways affecting VC. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed on the DEGs. Meanwhile, using the String database and Cytoscape software to construct protein-protein interaction (PPI) networks and identify hub genes with the highest module scores. Correlation analysis was performed for hub genes. Receiver operating characteristic (ROC) curves, expression level analysis, GSEA, and subcellular localization were performed for each hub gene. Expression of hub genes in normal and calcified vascular tissues was verified by quantitative reverse transcription PCR (RT-qPCR) and immunohistochemistry (IHC) experiments. The hub gene-related miRNA-mRNA and TF-mRNA networks were constructed and functionally enriched for analysis. Finally, the DGIdb database was utilized to search for alternative drugs targeting VC hub genes. Results By comparing the genes with normal vessels, there were 64 DEGs in mildly calcified vessels and 650 DEGs in severely calcified vessels. Spp1, Sost, Col1a1, Fn1, and Ibsp were central in the progression of the entire VC by the MCODE plug-in. These hub genes are primarily enriched in ossification, extracellular matrix, and ECM-receptor interactions. Expression level results showed that Spp1, Sost, Ibsp, and Fn1 were significantly highly expressed in VC, and Col1a1 was incredibly low. RT-qPCR and IHC validation results were consistent with bioinformatic analysis. We found multiple pathways of hub genes acting in VC and identified 16 targeting drugs. Conclusions This study perfected the molecular regulatory mechanism of VC. Our results indicated that Spp1, Sost, Col1a1, Fn1, and Ibsp could be potential novel biomarkers for VC and promising therapeutic targets.
Collapse
Affiliation(s)
- Chuanzhen Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Yinteng Wu
- Department of Orthopedic and Trauma Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Hai-lin Lu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Kai Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Xiao Qin
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| |
Collapse
|
9
|
Bouderlique E, Nollet L, Letavernier E, Vanakker OM. Minocycline Counteracts Ectopic Calcification in a Murine Model of Pseudoxanthoma Elasticum: A Proof-of-Concept Study. Int J Mol Sci 2022; 23:ijms23031838. [PMID: 35163765 PMCID: PMC8837001 DOI: 10.3390/ijms23031838] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 02/06/2023] Open
Abstract
Pseudoxanthoma elasticum (PXE) is an intractable Mendelian disease characterized by ectopic calcification in skin, eyes and blood vessels. Recently, increased activation of the DNA damage response (DDR) was shown to be involved in PXE pathogenesis, while the DDR/PARP1 inhibitor minocycline was found to attenuate aberrant mineralization in PXE cells and zebrafish. In this proof-of-concept study, we evaluated the anticalcifying properties of minocycline in Abcc6−/− mice, an established mammalian PXE model. Abcc6−/− mice received oral minocycline supplementation (40 mg/kg/day) from 12 to 36 weeks of age and were compared to untreated Abcc6−/− and Abcc6+/+ siblings. Ectopic calcification was evaluated using X-ray microtomography with three-dimensional reconstruction of calcium deposits in muzzle skin and Yasue’s calcium staining. Immunohistochemistry for the key DDR marker H2AX was also performed. Following minocycline treatment, ectopic calcification in Abcc6−/− mice was significantly reduced (−43.4%, p < 0.0001) compared to untreated Abcc6−/− littermates. H2AX immunostaining revealed activation of the DDR at sites of aberrant mineralization in untreated Abcc6−/− animals. In conclusion, we validated the anticalcifying effect of minocycline in Abcc6−/− mice for the first time. Considering its favorable safety profile in humans and low cost as a generic drug, minocycline may be a promising therapeutic compound for PXE patients.
Collapse
Affiliation(s)
- Elise Bouderlique
- UMR S 1155, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Université, 75020 Paris, France; (E.B.); (E.L.)
| | - Lukas Nollet
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium;
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Ectopic Mineralization Research Group, 9000 Ghent, Belgium
| | - Emmanuel Letavernier
- UMR S 1155, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Université, 75020 Paris, France; (E.B.); (E.L.)
| | - Olivier M. Vanakker
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium;
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Ectopic Mineralization Research Group, 9000 Ghent, Belgium
- Correspondence:
| |
Collapse
|
10
|
Shen M, Jiao K, Wang C, Ehrlich H, Wan M, Hao D, Li J, Wan Q, Tonggu L, Yan J, Wang K, Ma Y, Chen J, Tay FR, Niu L. Extracellular DNA: A Missing Link in the Pathogenesis of Ectopic Mineralization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103693. [PMID: 34939364 PMCID: PMC8844461 DOI: 10.1002/advs.202103693] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/10/2021] [Indexed: 05/12/2023]
Abstract
Although deoxyribonucleic acid (DNA) is the genetic coding for the very essence of life, these macromolecules or components thereof are not necessarily lost after a cell dies. There appears to be a link between extracellular DNA and biomineralization. Here the authors demonstrate that extracellular DNA functions as an initiator of collagen intrafibrillar mineralization. This is confirmed with in vitro and in vivo biological mineralization models. Because of their polyanionic property, extracellular DNA molecules are capable of stabilizing supersaturated calcium phosphate solution and mineralizing 2D and 3D collagen matrices completely as early as 24 h. The effectiveness of extracellular DNA in biomineralization of collagen is attributed to the relatively stable formation of amorphous liquid droplets triggered by attraction of DNA to the collagen fibrils via hydrogen bonding. These findings suggest that extracellular DNA is biomimetically significant for fabricating inorganic-organic hybrid materials for tissue engineering. DNA-induced collagen intrafibrillar mineralization provides a clue to the pathogenesis of ectopic mineralization in different body tissues. The use of DNase for targeting extracellular DNA at destined tissue sites provides a potential solution for treatment of diseases associated with ectopic mineralization.
Collapse
Affiliation(s)
- Min‐juan Shen
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Kai Jiao
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Chen‐yu Wang
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Hermann Ehrlich
- Institute of Electronic and Sensor MaterialsTU Bergakademie FreibergFreiberg09599Germany
- Center for Advanced TechnologyAdam Mickiewicz UniversityPoznan61‐614Poland
| | - Mei‐chen Wan
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Dong‐xiao Hao
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- Department of Applied PhysicsXi'an Jiaotong UniversityXi'anShaanxi710049P. R. China
| | - Jing Li
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Qian‐qian Wan
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Lige Tonggu
- School of MedicineUniversity of WashingtonSeattleWA98195USA
| | - Jian‐fei Yan
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Kai‐yan Wang
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Yu‐xuan Ma
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Ji‐hua Chen
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Franklin R. Tay
- The Dental College of GeorgiaAugusta UniversityAugustaGA30912USA
| | - Li‐na Niu
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology & Shaanxi Key Laboratory of Stomatology, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| |
Collapse
|
11
|
Herrmann J, Xia M, Gummi MR, Greco A, Schacke A, van der Giet M, Tölle M, Schuchardt M. Stressor-Induced "Inflammaging" of Vascular Smooth Muscle Cells via Nlrp3-Mediated Pro-inflammatory Auto-Loop. Front Cardiovasc Med 2022; 8:752305. [PMID: 34988126 PMCID: PMC8720922 DOI: 10.3389/fcvm.2021.752305] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/29/2021] [Indexed: 01/14/2023] Open
Abstract
Calcification of the vessel wall as one structural pathology of aged vessels is associated with high cardiovascular mortality of elderly patients. Aging is linked to chronic sterile inflammation and high burden of reactive oxygen species (ROS), leading to activation of pattern recognition receptors (PRRs) such as Nlrp3 in vascular cells. The current study investigates the role of PRR activation in the calcification of vascular smooth muscle cells (VSMCs). Therefore, in vitro cell culture of primary rat VSMCs and ex vivo aortic stimulations were used to analyze osteogenic, senescence and inflammatory markers via real-time PCR, in situ RNA hybridization, Western Blot, photometric assays and histological staining. Induction of ROS and DNA-damage by doxorubicin induces a shift of VSMC phenotype toward the expression of osteogenic, senescence and inflammatory proteins. Induction of calcification is dependent on Nlrp3 activity. Il-1β as a downstream target of Nlrp3 induces the synthetic, pro-calcifying VSMC phenotype. Inhibition of PRR with subsequent reduction of chronic inflammation might be an interesting target for reduction of calcification of VSMCs, with subsequent reduction of cardiovascular mortality of patients suffering from vessel stiffness.
Collapse
Affiliation(s)
- Jaqueline Herrmann
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany.,Department of Chemistry, Biochemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Mengdi Xia
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany.,Department of Nephrology, Second Clinical Medical Institution of North Sichuan Medical College (Nanchong Central Hospital), Nanchong, China
| | - Manasa Reddy Gummi
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| | - Anna Greco
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| | - Annika Schacke
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| | - Markus van der Giet
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| | - Markus Tölle
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| | - Mirjam Schuchardt
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| |
Collapse
|
12
|
Nollet L, Van Gils M, Willaert A, Coucke PJ, Vanakker OM. Minocycline attenuates excessive DNA damage response and reduces ectopic calcification in pseudoxanthoma elasticum. J Invest Dermatol 2021; 142:1629-1638.e6. [PMID: 34742705 DOI: 10.1016/j.jid.2021.10.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/27/2021] [Accepted: 10/19/2021] [Indexed: 12/25/2022]
Abstract
Pseudoxanthoma elasticum (PXE) is a hereditary ectopic calcification disorder affecting the skin, eyes and blood vessels. Recently, the DNA damage response (DDR), in particular poly(ADP-ribose) polymerase 1 (PARP1), was shown to be involved in aberrant mineralization raising the hypothesis that excessive DDR/PARP1 signaling also contributes to PXE pathogenesis. Using PXE patient and control fibroblasts, (lesional) skin tissue and abcc6a-/- zebrafish, we performed expression analysis of DDR/PARP1 targets with QRT-PCR, western blot, immunohistochemistry and enzyme activity assays; before and after treatment with the PARP1 inhibitor minocycline. PARP1 and the ATM-p21-p53 axis was found to be significantly increased in PXE. Additionally, PARP1 downstream targets IL-6, STAT1/3, TET1 and RUNX2 were upregulated while the RUNX2-antagonist microRNA-204 was decreased. In PXE fibroblasts, DDR/PARP1 signaling increased with advancing ectopic calcification. Minocycline treatment attenuated DDR/PARP1 overexpression and reduced aberrant mineralization in PXE fibroblasts and abcc6a-/- zebrafish. In summary, we demonstrated the involvement of excessive DDR/PARP1 signaling in PXE pathophysiology, identifying a STAT-driven cascade resulting in increased expression of the epigenetic modifier TET1 and pro-calcifying transcription factor RUNX2. Minocycline attenuated this deleterious molecular mechanism and reduced ectopic calcification both in vitro and in vivo, fueling the exciting prospect of a novel therapeutic compound for PXE.
Collapse
Affiliation(s)
- Lukas Nollet
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Ectopic Mineralization Research Group Ghent, Ghent, Belgium
| | - Matthias Van Gils
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Ectopic Mineralization Research Group Ghent, Ghent, Belgium
| | - Andy Willaert
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Paul J Coucke
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Olivier M Vanakker
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Ectopic Mineralization Research Group Ghent, Ghent, Belgium.
| |
Collapse
|
13
|
Whitehead M, Shanahan CM. Circulating uromodulin: a cytokine trap for osteoinductive inflammatory mediators in chronic kidney disease? Cardiovasc Res 2021; 117:651-652. [PMID: 33367494 DOI: 10.1093/cvr/cvaa348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Meredith Whitehead
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Catherine M Shanahan
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
14
|
Matuszyk E, Adamczyk A, Radwan B, Pieczara A, Szcześniak P, Mlynarski J, Kamińska K, Baranska M. Multiplex Raman imaging of organelles in endothelial cells. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 255:119658. [PMID: 33744837 DOI: 10.1016/j.saa.2021.119658] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 06/12/2023]
Abstract
Raman imaging using molecular reporters is a relatively new approach in subcellular investigations. It enables the visualization of organelles in cells with better selectivity and sensitivity compared to the label-free approach. Essentially Raman reporters possess in their structure an alkyne molecular group that can be selectively identified in the spectral region silent for biomolecules, hence facilitate the localization of individual organelles. The aim of this work is to visualize the main cell organelles in endothelial cells (HMEC-1) using established reporters (EdU and MitoBADY), but also to test a new one, namely falcarinol, which exhibits lipophilic properties. Moreover, we tested the possibility to use Raman reporters as a probe to detect changes in distribution of certain organelles after induced endothelial dysfunction (ED) in in vitro models. In both cases, induced ED is characterized by the formation of lipid droplets in the cells, which is why a good tool for the detection of lipid-rich organelles is so important in these studies. Two-dimensional Raman images were obtained, visualizing the distribution of selected organic compounds in the cell, such as proteins, lipids, and nucleic acids. Additionally, the distribution of EdU, MitoBADY and falcarinol in endothelial cells (ECs) was determined. Moreover, we highlight some drawback of established Raman reporter and the need for testing them in various physiological state of the cell.
Collapse
Affiliation(s)
- Ewelina Matuszyk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland.
| | - Adriana Adamczyk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Basseem Radwan
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Anna Pieczara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Piotr Szcześniak
- Institute of Organic Chemistry, Polish Academy of Sciences, 44/52 Kasprzaka Str., 01-224 Warsaw, Poland
| | - Jacek Mlynarski
- Institute of Organic Chemistry, Polish Academy of Sciences, 44/52 Kasprzaka Str., 01-224 Warsaw, Poland
| | - Katarzyna Kamińska
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Malgorzata Baranska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland.
| |
Collapse
|
15
|
Jianfeng W, Yutao W, Jianbin B. Long non-coding RNAs correlate with genomic stability in prostate cancer: A clinical outcome and survival analysis. Genomics 2021; 113:3141-3151. [PMID: 34174340 DOI: 10.1016/j.ygeno.2021.06.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/30/2021] [Accepted: 06/21/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) participate in the regulation of genomic stability. Understanding their biological functions can help us identify the mechanisms of the occurrence and progression of cancers and can provide theoretical guidance and the basis for treatment. RESULTS Based on the mutation hypothesis, we proposed a computational framework to identify genomic instability-related lncRNAs. Based on the differentially-expressed lncRNAs (DElncRNAs), we constructed a genomic instability-derived lncRNA signature (GILncSig) to calculate and stratify outcomes in patients with prostate cancer. It is an independent predictor of overall survival. The area under the curve = 0.805. This value may be more significant than the classic prognostic markers TP53 and Speckle-type POZ protein (SPOP) in terms of outcome prediction. CONCLUSIONS In summary, we conducted a computation approach and resource for mining genome instability-related lncRNAs. It may turn out to be highly significant for genomic instability and customized decision-making for patients with prostate cancer. It also may lead to effective methods and resources to study the molecular mechanism of genomic instability-related lncRNAs.
Collapse
Affiliation(s)
- Wang Jianfeng
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Wang Yutao
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Bi Jianbin
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
16
|
Sjakste N, Riekstiņa U. DNA damage and repair in differentiation of stem cells and cells of connective cell lineages: A trigger or a complication? Eur J Histochem 2021; 65. [PMID: 33942598 PMCID: PMC8116775 DOI: 10.4081/ejh.2021.3236] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023] Open
Abstract
The review summarizes literature data on the role of DNA breaks and DNA repair in the differentiation of pluripotent stem cells (PSC) and connective cell lineages. PSC, including embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC), are rapidly dividing cells with highly active DNA damage response (DDR) mechanisms to ensure the stability and integrity of the DNA. In PSCs, the most common DDR mechanism is error-free homologous recombination (HR) that is primarily active during the S phase of the cell cycle, whereas in quiescent, slow-dividing or non-dividing tissue progenitors and terminally differentiated cells, errorprone non-homologous end joining (NHEJ) mechanism of the double-strand break (DSB) repair is dominating. Thus, it seems that reprogramming and differentiation induce DNA strand breaks in stem cells which itself may trigger the differentiation process. Somatic cell reprogramming to iPSCs is preceded by a transient increase of the DSBs induced presumably by the caspase-dependent DNase or reactive oxygen species. In general, pluripotent stem cells possess stronger DNA repair systems compared to differentiated cells. Nonetheless, during a prolonged cell culture propagation, DNA breaks can accumulate due to the DNA polymerase stalling. Consequently, the DNA damage might trigger the differentiation of stem cells or replicative senescence of somatic cells. The differentiation process per se is often accompanied by a decrease in the DNA repair capacity. Thus, the differentiation might be triggered by DNA breaks, alternatively, the breaks can be a consequence of the decay in the DNA repair capacity of differentiated cells.
Collapse
|
17
|
Adamczyk A, Matuszyk E, Radwan B, Rocchetti S, Chlopicki S, Baranska M. Toward Raman Subcellular Imaging of Endothelial Dysfunction. J Med Chem 2021; 64:4396-4409. [PMID: 33821652 PMCID: PMC8154563 DOI: 10.1021/acs.jmedchem.1c00051] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
![]()
Multiple diseases are at some point associated with altered endothelial
function, and endothelial dysfunction (ED) contributes to their pathophysiology.
Biochemical changes of the dysfunctional endothelium are linked to
various cellular organelles, including the mitochondria, endoplasmic
reticulum, and nucleus, so organelle-specific insight is needed for
better understanding of endothelial pathobiology. Raman imaging, which
combines chemical specificity with microscopic resolution, has proved
to be useful in detecting biochemical changes in ED at the cellular
level. However, the detection of spectroscopic markers associated
with specific cell organelles, while desirable, cannot easily be achieved
by Raman imaging without labeling. This critical review summarizes
the current advances in Raman-based analysis of ED, with a focus on
a new approach involving molecular Raman reporters that could facilitate
the study of biochemical changes in cellular organelles. Finally,
imaging techniques based on both conventional spontaneous Raman scattering
and the emerging technique of stimulated Raman scattering are discussed.
Collapse
Affiliation(s)
- Adriana Adamczyk
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland.,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Ewelina Matuszyk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Basseem Radwan
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland.,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Stefano Rocchetti
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland.,Chair of Pharmacology, Jagiellonian University, 16 Grzegorzecka Str., 31-531 Krakow, Poland
| | - Malgorzata Baranska
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland.,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| |
Collapse
|
18
|
Chandra A, Rajawat J. Skeletal Aging and Osteoporosis: Mechanisms and Therapeutics. Int J Mol Sci 2021; 22:ijms22073553. [PMID: 33805567 PMCID: PMC8037620 DOI: 10.3390/ijms22073553] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
Bone is a dynamic organ maintained by tightly regulated mechanisms. With old age, bone homeostasis, which is maintained by an intricate balance between bone formation and bone resorption, undergoes deregulation. Oxidative stress-induced DNA damage, cellular apoptosis, and cellular senescence are all responsible for this tissue dysfunction and the imbalance in the bone homeostasis. These cellular mechanisms have become a target for therapeutics to treat age-related osteoporosis. Genetic mouse models have shown the importance of senescent cell clearance in alleviating age-related osteoporosis. Furthermore, we and others have shown that targeting cellular senescence pharmacologically was an effective tool to alleviate age- and radiation-induced osteoporosis. Senescent cells also have an altered secretome known as the senescence associated secretory phenotype (SASP), which may have autocrine, paracrine, or endocrine function. The current review discusses the current and potential pathways which lead to a senescence profile in an aged skeleton and how bone homeostasis is affected during age-related osteoporosis. The review has also discussed existing therapeutics for the treatment of osteoporosis and rationalizes for novel therapeutic options based on cellular senescence and the SASP as an underlying pathogenesis of an aging bone.
Collapse
Affiliation(s)
- Abhishek Chandra
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA
- Department of Internal Medicine, Division of Geriatric Medicine and Gerontology, Mayo Clinic, Rochester, MN 55902, USA
- Robert and Arlene Kogod Aging Center, Mayo Clinic, Rochester, MN 55902, USA
- Correspondence: ; Tel.: +1-507-266-1847
| | - Jyotika Rajawat
- Department of Zoology, University of Lucknow, University Rd, Babuganj, Hasanganj, Lucknow, Uttar Pradesh 226007, India;
| |
Collapse
|