1
|
Gong W, Yu H, You W, Chen Z, Wang Y, Liu C, Li Y, Guan S. The oral microbiota: new insight into intracranial aneurysms. Ann Med 2025; 57:2451191. [PMID: 39803910 PMCID: PMC11731255 DOI: 10.1080/07853890.2025.2451191] [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: 07/28/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Intracranial aneurysms (IAs) are a significant clinical concern, with detection rates increasing due to advances in imaging technologies. However, precise mechanisms underlying their pathophysiology remain incompletely understood. Recent evidence suggests a pivotal role of oral microbiota dysbiosis, particularly periodontal pathogens, in systemic inflammation that may contribute to IA development and rupture. OBJECTIVE This review aims to critically evaluate the association between oral microbiota dysbiosis and the pathogenesis of IAs, with a focus on the molecular and immunological mechanisms by which oral pathogens influence vascular pathology. METHODS We conducted a comprehensive analysis of the literature regarding the impact of oral microbial dysbiosis on IA pathophysiology, emphasizing the role of specific pathogenic species, such as Porphyromonas gingivalis. The review explores how these pathogens may mediate chronic inflammation through hematogenous spread, gut microbiome alterations, and neuroinflammatory processes, leading to vascular remodeling and cerebrovascular instability. RESULTS The findings suggest that oral microbial dysbiosis, particularly the presence of pathogenic bacteria, is implicated in the systemic inflammatory response that exacerbates the structural integrity of the cerebrovascular wall. Chronic inflammatory states induced by oral pathogens contribute to extracellular matrix degradation, impaired vascular remodeling, and an increased susceptibility to IA rupture. CONCLUSIONS The findings highlight the importance of maintaining oral microbiota homeostasis as a potential therapeutic target for preventing IAs. Interventions aimed at restoring oral microbial balance may represent a novel strategy for reducing the burden of IA formation and rupture, highlighting the need for an integrated approach to oral health and IAs prevention.
Collapse
Affiliation(s)
- Wentao Gong
- Department of Neurointervention, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hairong Yu
- Department of Neurology, Shidao People’s Hospital of Rongcheng, Weihai, China
| | - Wei You
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhen Chen
- Department of Neurointervention, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chao Liu
- Department of Neurointervention, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Youxiang Li
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Sheng Guan
- Department of Neurointervention, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Kailash KA, Akanda SR, Davis AL, Crandall CL, Zaghloul MS, Setton LA, Halabi CM, Zayed MA, Wagenseil JE. Transport across the thoracic aortic wall: implications for aneurysm pathobiology, diagnosis, and treatment. Am J Physiol Heart Circ Physiol 2025; 328:H1113-H1129. [PMID: 40192071 DOI: 10.1152/ajpheart.00886.2024] [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: 12/19/2024] [Revised: 01/03/2025] [Accepted: 03/19/2025] [Indexed: 05/01/2025]
Abstract
Thoracic aortic aneurysms (TAAs) are a dilation of the aorta that may fatally dissect or rupture. The current clinical management for TAA is continuous monitoring and surgical replacement once the aortic diameter reaches a specified size or rate of growth. Although operative intervention is often successful in preventing fatal outcomes, not all patients will reach surgical criteria before an aortic event, and the surgery carries significant risk with a potential requirement for reoperation. There is a need for patient-specific diagnostic tools and/or novel therapeutics to treat TAA. In this review, we discuss fluid and solute transport through the aortic wall (transmural aortic transport), its potential contributions to TAA progression, and possible applications for diagnosis and treatment. We first discuss the structural organization of the aortic wall with a focus on cellular and extracellular matrix (ECM) changes associated with TAA that may alter transmural transport. We then focus on aortic transmural transport processes defined with biphasic and multiphasic theory. Biphasic theory describes fluid interactions with a porous solid (i.e., the aortic wall), whereas multiphasic theory describes fluid and solute(s) interactions with a porous solid. We summarize experimental and computational methods to quantify transport through the aortic wall. Finally, we discuss how transmural transport may be used to diagnose, monitor, or treat TAA. Further understanding of transmural transport may lead to new insights into TAA pathobiology and future clinical solutions.
Collapse
Grants
- R01HL133662 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL164800 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL166448 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL172996 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL153262 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL153436 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL150891 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01AR0776780 HHS | NIH | National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
- Pharmaceutical Research and Manufacturers of America Foundation (PhRMAF)
Collapse
Affiliation(s)
- Keshav A Kailash
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Shamimur R Akanda
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Alexandra L Davis
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Christie L Crandall
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Mohamed S Zaghloul
- Department of Surgery, Washington University, St. Louis, Missouri, United States
| | - Lori A Setton
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
- Department of Orthopedic Surgery, Washington University, St. Louis, Missouri, United States
| | - Carmen M Halabi
- Department of Pediatrics, Washington University, St. Louis, Missouri, United States
| | - Mohamed A Zayed
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
- Department of Surgery, Washington University, St. Louis, Missouri, United States
- Department of Radiology, Washington University, St. Louis, Missouri, United States
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| |
Collapse
|
3
|
Dalal AR, Pedroza AJ, Kim J, Gilles C, Gu W, Kusadokoro S, Shad R, Mitchel O, Jackson W, Hiesinger W, Berry G, MacFarlane EG, Quertermous T, Cheng P, Fischbein MP. Chemokine (C-C Motif) Ligand 2 Expressing Adventitial Fibroblast Expansion During Loeys-Dietz Syndrome Aortic Aneurysm Formation. Arterioscler Thromb Vasc Biol 2025; 45:722-742. [PMID: 40109260 PMCID: PMC12018128 DOI: 10.1161/atvbaha.124.322069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Loeys-Dietz syndrome (LDS), caused by mutations in the TGF-β (transforming growth factor-β) signaling cascade, leads to aggressive thoracic aneurysms. While vascular smooth muscle cell (SMC) phenotype modulation has been implicated in thoracic aneurysm formation, we sought to characterize the role of cell state transitions in LDS aneurysm pathogenesis. METHODS We performed single-cell transcriptomic characterization of aortic root/ascending aorta from a murine LDS model (Tgfbr2G357W/+ versus littermate WT [wild-type] control) at 8 weeks, 24 weeks, and aortic root/ascending aortic samples from human LDS surgical specimens (n=5 LDS [TGFBR1/2] and n=2 donor control) to understand cell state transitions and transcriptomic alterations in LDS. Select cell markers were spatially localized with RNA in situ hybridization, immunofluorescence, and immunohistochemistry. Single-cell RNA sequencing of murine and human LDS samples (>30 000 cells) revealed unique SMC, fibroblast, and macrophage transcriptomic profiles in LDS. RESULTS Instead of SMC phenotypic modulation seen in Marfan syndrome, transcriptomic alterations observed in LDS are most prominent in the adventitial fibroblast in the Tgfbr2G357W/+ mouse model. While a distinct modulated SMC cluster does not appear in Tgfbr2G357W/+, SMCs transcriptomically differ from WT counterparts. Adventitial fibroblasts were activated into a proinflammatory state associated with increased macrophage recruitment (Ccl2, Il6, Ccl7, and Cxcl2) and fibrotic response genes (Col1a1, Col1a2, and Col3a1), with a 6-fold increase in aortic wall macrophage content in Tgfbr2G357W/+ compared with WT. Similar findings were also observed in human LDS aortic samples with increased proinflammatory adventitial fibroblast transcriptomic program in parallel with heightened macrophage recruitment. CONCLUSIONS Despite phenotypic similarities in aneurysm formation, the dominant cellular and molecular mechanism of Marfan syndrome and LDS aneurysms are distinct. LDS mouse and human adventitial fibroblasts transcriptomically modulate into a proinflammatory state. Adventitial fibroblasts, in addition to SMCs, are another important pathological cell population during LDS aneurysm formation to consider for targeted therapy to potentially impede LDS aneurysm formation.
Collapse
MESH Headings
- Animals
- Loeys-Dietz Syndrome/genetics
- Loeys-Dietz Syndrome/pathology
- Loeys-Dietz Syndrome/metabolism
- Loeys-Dietz Syndrome/complications
- Humans
- Adventitia/pathology
- Adventitia/metabolism
- Receptor, Transforming Growth Factor-beta Type II/genetics
- Receptor, Transforming Growth Factor-beta Type II/metabolism
- Disease Models, Animal
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Female
- Transcriptome
- Phenotype
- Macrophages/metabolism
- Macrophages/pathology
- Aortic Aneurysm/pathology
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/genetics
- Chemokine CCL2/metabolism
- Chemokine CCL2/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Aorta/pathology
- Aorta/metabolism
- Single-Cell Analysis
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
Collapse
Affiliation(s)
- Alex R. Dalal
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Jennifer Kim
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Casey Gilles
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Wenduo Gu
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford CA USA
| | - Sho Kusadokoro
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Rohan Shad
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Olivia Mitchel
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - William Jackson
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford CA USA
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Gerald Berry
- Department of Pathology, Stanford University School of Medicine, Stanford CA USA
| | - Elena Gallo MacFarlane
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford CA USA
| | - Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford CA USA
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| |
Collapse
|
4
|
Chen HN, Hu YN, Ran LL, Wang M, Zhang Z. Sexual dimorphism in aortic aneurysm: A review of the contributions of sex hormones and sex chromosomes. Vascul Pharmacol 2025; 158:107460. [PMID: 39716526 DOI: 10.1016/j.vph.2024.107460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/23/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Aortic aneurysm is a common cardiovascular disease. Over time, the disease damages the structural and functional integrity of the aorta, causing it to abnormally expand and potentially rupture, which can be fatal. Sex differences are evident in the disease, with men experiencing an earlier onset and higher incidence. However, women may face a worse prognosis and a higher risk of rupture. While there are some studies on the cellular and molecular mechanisms of aneurysm formation, it remains unclear how sex factors contribute to sexual dimorphism. Therefore, this review aims to summarize the role of sex in the occurrence of aortic aneurysms, offering valuable insights for disease prevention and the development of appropriate treatment options.
Collapse
Affiliation(s)
- Hao-Nan Chen
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Yan-Ni Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Li-Ling Ran
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Mi Wang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
5
|
Wu H, Yang X, Chen T, Yu B, Chen M, Wang T, Jiang L, Zhang B, Zhou X, Cheng J, Chen K, Zhang T, Hu Y, Xu S, Lian J, Zhang H, Xiao Q, Ye H, Xu Q. Aneurysm Is Restricted by CD34 + Cell-Formed Fibrous Collars Through the PDGFRb-PI3K Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408996. [PMID: 39731355 PMCID: PMC11831520 DOI: 10.1002/advs.202408996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/05/2024] [Indexed: 12/29/2024]
Abstract
Aortic aneurysm is a life-threatening disease caused by progressive dilation of the aorta and weakened aortic walls. Its pathogenesis involves an imbalance between connective tissue repair and degradation. CD34+ cells comprise a heterogeneous population that exhibits stem cell and progenitor cell properties. However, the role of CD34+ cells in abdominal aortic aneurysm (AAA) remains unclear. In this study, downregulated CD34 expression is observed in aneurysmal aortas from both patients and mouse models compared to that in non-dilated aortas. Furthermore, by combining Cd34-CreERT2;Rosa26-tdTomato;(Apoe-/-) lineage tracing, bone marrow transplantation, and single-cell sequencing, it is found that during AAA development, non-bone marrow CD34+ cells are activated to transdifferentiate into Periostin+ myofibroblasts, thereby contributing to the formation of fibrotic collars. Dual recombinase-based lineage tracing confirms the presence and involvement of CD34+/Periostin+ myofibroblasts in fibrotic collar formation during AAA development. Functionally, selective depletion of systemic or non-bone marrow CD34+ cells, as well as CD34+/Periostin+ myofibroblasts, by diphtheria toxin significantly exacerbates AAA progression and increases disease mortality. Mechanistically, it is identified that the PDGF-PDGFRb-PI3K axis is indispensable for Periostin+ myofibroblast generation from non-bone marrow CD34+ cells in AAA, offering a new therapeutic target for patients with AAA at a high risk of rupture.
Collapse
Affiliation(s)
- Hong Wu
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Xiaoping Yang
- Department of CardiologyNingbo Institute of Innovation for Combined Medicine and EngineeringLihuili Hospital Affiliated to Ningbo UniversityNingbo UniversityNingboZhejiang315000China
| | - Ting Chen
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Baoqi Yu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesCapital Medical UniversityKey Laboratory of Remodeling‐Related Cardiovascular DiseasesMinistry of EducationBeijing Key Laboratory of Metabolic Disorder‐Related Cardiovascular DiseasesBeijing100069China
| | - Mengjia Chen
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Ting Wang
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Liujun Jiang
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Bohuan Zhang
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Xuhao Zhou
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Junning Cheng
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Kai Chen
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Tao Zhang
- Department of Vascular SurgeryPeking University People's HospitalBeijing100044China
| | - Yanhua Hu
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Simon Xu
- Department of SurgeryLiverpool Heart and Chest HospitalLiverpoolL14 3PEUK
| | - Jiangfang Lian
- Department of CardiologyNingbo Institute of Innovation for Combined Medicine and EngineeringLihuili Hospital Affiliated to Ningbo UniversityNingbo UniversityNingboZhejiang315000China
| | - Hongkun Zhang
- Department of Vascular SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision MedicineWilliam Harvey Research InstituteFaculty of Medicine and DentistryQueen Mary University of LondonLondonEC1M 6BQUK
| | - Honghua Ye
- Department of CardiologyNingbo Institute of Innovation for Combined Medicine and EngineeringLihuili Hospital Affiliated to Ningbo UniversityNingbo UniversityNingboZhejiang315000China
| | - Qingbo Xu
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| |
Collapse
|
6
|
Stougiannou TM, Christodoulou KC, Karangelis D. Olfactory Receptors and Aortic Aneurysm: Review of Disease Pathways. J Clin Med 2024; 13:7778. [PMID: 39768700 PMCID: PMC11727755 DOI: 10.3390/jcm13247778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Aortic aneurysm, the pathological dilatation of the aorta at distinct locations, can be attributed to many different genetic and environmental factors. The resulting pathobiological disturbances generate a complex interplay of processes affecting cells and extracellular molecules of the tunica interna, media and externa. In short, aortic aneurysm can affect processes involving the extracellular matrix, lipid trafficking/atherosclerosis, vascular smooth muscle cells, inflammation, platelets and intraluminal thrombus formation, as well as various endothelial functions. Many of these processes are interconnected, potentiating one another. Newer discoveries, including the involvement of odorant olfactory receptors in these processes, have further shed light on disease initiation and pathology. Olfactory receptors are a varied group of G protein coupled-receptors responsible for the recognition of chemosensory information. Although they comprise many different subgroups, some of which are not well-characterized or identified in humans, odorant olfactory receptors, in particular, are most commonly associated with recognition of olfactory information. They can also be ectopically localized and thus carry out additional functions relevant to the tissue in which they are identified. It is thus the purpose of this narrative review to summarize and present pathobiological processes relevant to the initiation and propagation of aortic aneurysm, while also incorporating evidence associating these ectopically functioning odorant olfactory receptors with the overall pathology.
Collapse
Affiliation(s)
- Theodora M. Stougiannou
- Department of Cardiothoracic Surgery, University General Hospital, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.C.C.); (D.K.)
| | | | | |
Collapse
|
7
|
Teti G, Camiletti R, Gatta V, Longhin A, Falconi M. Vascular Mesenchymal Stromal Cells and Cellular Senescence: A Two-Case Study Investigating the Correlation Between an Inflammatory Microenvironment and Abdominal Aortic Aneurysm Development. Int J Mol Sci 2024; 25:12495. [PMID: 39684205 PMCID: PMC11641388 DOI: 10.3390/ijms252312495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
An abdominal aortic aneurysm (AAA) is described as a gradual and localized permanent expansion of the aorta resulting from the weakening of the vascular wall. The key aspects of AAA's progression are high proteolysis of the structural elements of the vascular wall, the depletion of vascular smooth muscle cells (VSMCs), and a chronic immunoinflammatory response. The pathological mechanisms underpinning the development of an AAA are complex and still unknown. At present, there are no successful drug treatments available that can slow the progression of an AAA or prevent the rupture of the aneurysmal vascular wall. Recently, it has been suggested that endothelial cellular senescence may be involved in vascular aging and vascular aging diseases, but there is no clear correlation between cellular senescence and AAAs. Therefore, the aim of this study was to identify the presence of senescent cells on the vascular wall of aneurysmatic abdominal aortas and to correlate their distribution with the morphological markers of AAAs. Pathological and healthy segments of abdominal aortas were collected during repair surgery and immediately processed for histological and immunohistochemical analyses. Hematoxylin/eosin, Verhoeff-van Gieson, and Goldner's Masson trichrome staining procedures were carried out to investigate the morphological features related to the pathology. Immunohistochemical investigations for the p21cip1/waf1, p53, and NFkB markers were carried out to selectively identify positive cells in the vascular wall of the AAA samples related to cellular senescence and an inflammatory microenvironment. The results revealed the presence of a few senescent vascular cells on the aneurysmatic wall of the abdominal aortas, surrounded by a highly inflamed microenvironment that was highly expressed in the tunica media and adventitia of both pathological and healthy segments. Our data demonstrate the presence of senescent vascular cells in AAA samples, which could enhance the promotion of a high inflammatory vascular microenvironment, supporting the evolution of the pathology. Although this study was based on only two cases, the results highlight the importance of targeting cellular senescence to reduce an inflammatory microenvironment, which can support the progression of age-related diseases.
Collapse
Affiliation(s)
- Gabriella Teti
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (R.C.); (V.G.); (A.L.)
| | - Riccardo Camiletti
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (R.C.); (V.G.); (A.L.)
| | - Valentina Gatta
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (R.C.); (V.G.); (A.L.)
| | - Aurora Longhin
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (R.C.); (V.G.); (A.L.)
| | - Mirella Falconi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
8
|
Xie X, Wang P, Jin M, Wang Y, Qi L, Wu C, Guo S, Li C, Zhang X, Yuan Y, Ma X, Liu F, Liu W, Liu H, Duan C, Ye P, Li X, Borish L, Zhao W, Feng X. IL-1β-induced epithelial cell and fibroblast transdifferentiation promotes neutrophil recruitment in chronic rhinosinusitis with nasal polyps. Nat Commun 2024; 15:9101. [PMID: 39438439 PMCID: PMC11496833 DOI: 10.1038/s41467-024-53307-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
Neutrophilic inflammation contributes to multiple chronic inflammatory airway diseases, including asthma and chronic rhinosinusitis with nasal polyps (CRSwNP), and is associated with an unfavorable prognosis. Here, using single-cell RNA sequencing (scRNA-seq) to profile human nasal mucosa obtained from the inferior turbinates, middle turbinates, and nasal polyps of CRSwNP patients, we identify two IL-1 signaling-induced cell subsets-LY6D+ club cells and IDO1+ fibroblasts-that promote neutrophil recruitment by respectively releasing S100A8/A9 and CXCL1/2/3/5/6/8 into inflammatory regions. IL-1β, a pro-inflammatory cytokine involved in IL-1 signaling, induces the transdifferentiation of LY6D+ club cells and IDO1+ fibroblasts from primary epithelial cells and fibroblasts, respectively. In an LPS-induced neutrophilic CRSwNP mouse model, blocking IL-1β activity with a receptor antagonist significantly reduces the numbers of LY6D+ club cells and IDO1+ fibroblasts and mitigates nasal inflammation. This study implicates the function of two cell subsets in neutrophil recruitment and demonstrates an IL-1-based intervention for mitigating neutrophilic inflammation in CRSwNP.
Collapse
Affiliation(s)
- Xinyu Xie
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Medical and Health Discipline, Qilu Hospital of Shandong University, Jinan, China
| | - Pin Wang
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Medical and Health Discipline, Qilu Hospital of Shandong University, Jinan, China
| | - Min Jin
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yue Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Lijie Qi
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Medical and Health Discipline, Qilu Hospital of Shandong University, Jinan, China
| | - Changhua Wu
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
| | - Shu Guo
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
| | - Changqing Li
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaojun Zhang
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Medical and Health Discipline, Qilu Hospital of Shandong University, Jinan, China
| | - Ye Yuan
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinyi Ma
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
| | - Fangying Liu
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
| | - Weiyuan Liu
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
| | - Heng Liu
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
| | - Chen Duan
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Medical and Health Discipline, Qilu Hospital of Shandong University, Jinan, China
| | - Ping Ye
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Medical and Health Discipline, Qilu Hospital of Shandong University, Jinan, China
| | - Xuezhong Li
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Medical and Health Discipline, Qilu Hospital of Shandong University, Jinan, China
| | - Larry Borish
- Departments of Medicine, University of Virginia Health System, Charlottesville, VA, USA
- Departments of Microbiology, University of Virginia Health System, Charlottesville, VA, USA
| | - Wei Zhao
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, China
- Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong University, Jinan, China
| | - Xin Feng
- Department of Otorhinolaryngology, National Health Commission Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China.
- Shandong Provincial Key Medical and Health Discipline, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
9
|
Naegeli KM, Niklason LE. Engineered vascular grafts lend unique insight to pathophysiology of aortic aneurysms. Cell Stem Cell 2024; 31:1099-1100. [PMID: 39094540 DOI: 10.1016/j.stem.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Yang et al.1 generate tissue engineered blood vessels from hiPSC-derived smooth muscle cells harboring a mutation found in Loeys-Dietz syndrome. In vitro and in vivo data from these vessels provide new insight into the molecular physiology of aortic aneurysms and may create a paradigm for understanding a suite of vascular diseases.
Collapse
|
10
|
Macchioni P, Germanò G, Girolimetto N, Klinowski G, Gavioli L, Muratore F, Laneri A, Ricordi C, Marvisi C, Magnani L, Salvarani C. Ultrasound Examination of Common Carotid Adventitial Thickness Can Differentiate Takayasu Arteritis and Large Vessel Giant Cell Arteritis. J Pers Med 2024; 14:627. [PMID: 38929848 PMCID: PMC11205024 DOI: 10.3390/jpm14060627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/24/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Pathological studies have demonstrated that the adventitial layer is markedly thickened in Takayasu (TAK) as compared to large vessel giant cell arteritis (LV-GCA). An ultrasound (US) examination of the arterial vessels allows the determination of intima media thickness (IMT) and of adventitial layer thickness (extra media thickness (EMT)). No previous study has evaluated if there are differences in EMT thickness between TAK and LV-GCA. In this cross-sectional retrospective study of stored ultrasound (US) imaging, we have compared common carotid artery (CCA) EMT and IMT in a series of consecutive TAK and LV-GCA patients. US examination CCA IMT and EMT were significantly higher in TAK as compared to LV-GCA. With ROC curve analysis, we have found that an EMT > 0.76 mm has high sensitivity and specificity for TAK CCA examination. The percentage of CCA at EMT > 0.76 mm and the total arterial wall thickening were significantly higher in TAK group examinations. EMT thickness correlated with disease duration and IMT in the TAK group, as well as with the IMT and ESR values in the LV-GCA group. Upon multivariate logistic regression analysis, factors independently associated with TAK CCA were EMT > 0.76 mm and age. No significant variation in IMT and EMT could be demonstrated in subsequent US CCA examinations.
Collapse
Affiliation(s)
- Pierluigi Macchioni
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
| | - Giuseppe Germanò
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
| | - Nicolò Girolimetto
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
| | - Giulia Klinowski
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
| | - Letizia Gavioli
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
| | - Francesco Muratore
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Alessia Laneri
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
| | - Caterina Ricordi
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Chiara Marvisi
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Luca Magnani
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
| | - Carlo Salvarani
- Division of Rheumatology, Arcispedale Santa Maria Nuova, IRCCS, 42123 Reggio Emilia, Italy; (G.G.); (N.G.); (G.K.); (L.G.); (F.M.); (A.L.); (C.M.); (L.M.); (C.S.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| |
Collapse
|
11
|
Wagenhäuser MU, Mulorz J, Krott KJ, Bosbach A, Feige T, Rhee YH, Chatterjee M, Petzold N, Böddeker C, Ibing W, Krüger I, Popovic AM, Roseman A, Spin JM, Tsao PS, Schelzig H, Elvers M. Crosstalk of platelets with macrophages and fibroblasts aggravates inflammation, aortic wall stiffening, and osteopontin release in abdominal aortic aneurysm. Cardiovasc Res 2024; 120:417-432. [PMID: 37976180 DOI: 10.1093/cvr/cvad168] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/01/2023] [Accepted: 09/23/2023] [Indexed: 11/19/2023] Open
Abstract
AIMS Abdominal aortic aneurysm (AAA) is a highly lethal disease with progressive dilatation of the abdominal aorta accompanied by degradation and remodelling of the vessel wall due to chronic inflammation. Platelets play an important role in cardiovascular diseases, but their role in AAA is poorly understood. METHODS AND RESULTS The present study revealed that platelets play a crucial role in promoting AAA through modulation of inflammation and degradation of the extracellular matrix (ECM). They are responsible for the up-regulation of SPP1 (osteopontin, OPN) gene expression in macrophages and aortic tissue, which triggers inflammation and remodelling and also platelet adhesion and migration into the abdominal aortic wall and the intraluminal thrombus (ILT). Further, enhanced platelet activation and pro-coagulant activity result in elevated gene expression of various cytokines, Mmp9 and Col1a1 in macrophages and Il-6 and Mmp9 in fibroblasts. Enhanced platelet activation and pro-coagulant activity were also detected in AAA patients. Further, we detected platelets and OPN in the vessel wall and in the ILT of patients who underwent open repair of AAA. Platelet depletion in experimental murine AAA reduced inflammation and ECM remodelling, with reduced elastin fragmentation and aortic diameter expansion. Of note, OPN co-localized with platelets, suggesting a potential role of OPN for the recruitment of platelets into the ILT and the aortic wall. CONCLUSION In conclusion, our data strongly support the potential relevance of anti-platelet therapy to reduce AAA progression and rupture in AAA patients.
Collapse
Affiliation(s)
- Markus U Wagenhäuser
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Joscha Mulorz
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Kim J Krott
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Agnes Bosbach
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Tobias Feige
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Yae H Rhee
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Madhumita Chatterjee
- Department of Pharmacology, Experimental Therapy and Toxicology, University Hospital Tübingen, Wilhelmstrasse 5, 72074 Tübingen, Germany
| | - Niklas Petzold
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Christopher Böddeker
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Wiebke Ibing
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Irena Krüger
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Ana M Popovic
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Ann Roseman
- VA Palo Alto Health Care System, Palo Alto, 3801 Miranda Avenue, 94304 CA, USA
| | - Joshua M Spin
- VA Palo Alto Health Care System, Palo Alto, 3801 Miranda Avenue, 94304 CA, USA
- Department of Cardiovascular Medicine, Stanford University, 291 Campus Drive Stanford, 94305 CA, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto, 3801 Miranda Avenue, 94304 CA, USA
- Department of Cardiovascular Medicine, Stanford University, 291 Campus Drive Stanford, 94305 CA, USA
| | - Hubert Schelzig
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Margitta Elvers
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| |
Collapse
|
12
|
Alexander KC, Anderson CW, Agala CB, Tasoudis P, Collins EN, Ding Y, Blackwell JW, Willcox DE, Farivar BS, Kibbe MR, Ikonomidis JS, Akerman AW. Paradoxical Changes: EMMPRIN Tissue and Plasma Levels in Marfan Syndrome-Related Thoracic Aortic Aneurysms. J Clin Med 2024; 13:1548. [PMID: 38541774 PMCID: PMC10970932 DOI: 10.3390/jcm13061548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 11/03/2024] Open
Abstract
Background: Thoracic aortic aneurysms (TAAs) associated with Marfan syndrome (MFS) are unique in that extracellular matrix metalloproteinase inducer (EMMPRIN) levels do not behave the way they do in other cardiovascular pathologies. EMMPRIN is shed into the circulation through the secretion of extracellular vesicles. This has been demonstrated to be dependent upon the Membrane Type-1 MMP (MT1-MMP). We investigated this relationship in MFS TAA tissue and plasma to discern why unique profiles may exist. Methods: Protein targets were measured in aortic tissue and plasma from MFS patients with TAAs and were compared to healthy controls. The abundance and location of MT1-MMP was modified in aortic fibroblasts and secreted EMMPRIN was measured in conditioned culture media. Results: EMMPRIN levels were elevated in MFS TAA tissue but reduced in plasma, compared to the controls. Tissue EMMPRIN elevation did not induce MMP-3, MMP-8, or TIMP-1 expression, while MT1-MMP and TIMP-2 were elevated. MMP-2 and MMP-9 were reduced in TAA tissue but increased in plasma. In aortic fibroblasts, EMMPRIN secretion required the internalization of MT1-MMP. Conclusions: In MFS, impaired EMMPRIN secretion likely contributes to higher tissue levels, influenced by MT1-MMP cellular localization. Low EMMPRIN levels, in conjunction with other MMP analytes, distinguished MFS TAAs from controls, suggesting diagnostic potential.
Collapse
Affiliation(s)
- Kyle C. Alexander
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (K.C.A.); (C.B.A.); (Y.D.); (J.W.B.); (J.S.I.)
| | - Carlton W. Anderson
- Advanced Analytics Core, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Chris B. Agala
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (K.C.A.); (C.B.A.); (Y.D.); (J.W.B.); (J.S.I.)
| | - Panagiotis Tasoudis
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (K.C.A.); (C.B.A.); (Y.D.); (J.W.B.); (J.S.I.)
| | - Elizabeth N. Collins
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (K.C.A.); (C.B.A.); (Y.D.); (J.W.B.); (J.S.I.)
| | - Yiwen Ding
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (K.C.A.); (C.B.A.); (Y.D.); (J.W.B.); (J.S.I.)
| | - John W. Blackwell
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (K.C.A.); (C.B.A.); (Y.D.); (J.W.B.); (J.S.I.)
| | - Danielle E. Willcox
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (K.C.A.); (C.B.A.); (Y.D.); (J.W.B.); (J.S.I.)
| | - Behzad S. Farivar
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA (M.R.K.)
| | - Melina R. Kibbe
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA (M.R.K.)
- Department of Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - John S. Ikonomidis
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (K.C.A.); (C.B.A.); (Y.D.); (J.W.B.); (J.S.I.)
| | - Adam W. Akerman
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (K.C.A.); (C.B.A.); (Y.D.); (J.W.B.); (J.S.I.)
| |
Collapse
|
13
|
Singh AA, Shetty DK, Jacob AG, Bayraktar S, Sinha S. Understanding genomic medicine for thoracic aortic disease through the lens of induced pluripotent stem cells. Front Cardiovasc Med 2024; 11:1349548. [PMID: 38440211 PMCID: PMC10910110 DOI: 10.3389/fcvm.2024.1349548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/31/2024] [Indexed: 03/06/2024] Open
Abstract
Thoracic aortic disease (TAD) is often silent until a life-threatening complication occurs. However, genetic information can inform both identification and treatment at an early stage. Indeed, a diagnosis is important for personalised surveillance and intervention plans, as well as cascade screening of family members. Currently, only 20% of heritable TAD patients have a causative mutation identified and, consequently, further advances in genetic coverage are required to define the remaining molecular landscape. The rapid expansion of next generation sequencing technologies is providing a huge resource of genetic data, but a critical issue remains in functionally validating these findings. Induced pluripotent stem cells (iPSCs) are patient-derived, reprogrammed cell lines which allow mechanistic insights, complex modelling of genetic disease and a platform to study aortic genetic variants. This review will address the need for iPSCs as a frontline diagnostic tool to evaluate variants identified by genomic discovery studies and explore their evolving role in biological insight through to drug discovery.
Collapse
Affiliation(s)
| | | | | | | | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, United Kingdom
| |
Collapse
|
14
|
Carrabba M, Fagnano M, Ghorbel MT. Development of a Novel Hierarchically Biofabricated Blood Vessel Mimic Decorated with Three Vascular Cell Populations for the Reconstruction of Small-Diameter Arteries. ADVANCED FUNCTIONAL MATERIALS 2024; 34:adfm.202300621. [PMID: 39257639 PMCID: PMC7616429 DOI: 10.1002/adfm.202300621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Indexed: 09/12/2024]
Abstract
The availability of grafts to replace small-diameter arteries remains an unmet clinical need. Here, the validated methodology is reported for a novel hybrid tissue-engineered vascular graft that aims to match the natural structure of small-size arteries. The blood vessel mimic (BVM) comprises an internal conduit of co-electrospun gelatin and polycaprolactone (PCL) nanofibers (corresponding to the tunica intima of an artery), reinforced by an additional layer of PCL aligned fibers (the internal elastic membrane). Endothelial cells are deposited onto the luminal surface using a rotative bioreactor. A bioprinting system extrudes two concentric cell-laden hydrogel layers containing respectively vascular smooth muscle cells and pericytes to create the tunica media and adventitia. The semi-automated cellularization process reduces the production and maturation time to 6 days. After the evaluation of mechanical properties, cellular viability, hemocompatibility, and suturability, the BVM is successfully implanted in the left pulmonary artery of swine. Here, the BVM showed good hemostatic properties, capability to withstand blood pressure, and patency at 5 weeks post-implantation. These promising data open a new avenue to developing an artery-like product for reconstructing small-diameter blood vessels.
Collapse
Affiliation(s)
- Michele Carrabba
- Bristol Heart Institute, School of Translational Health Sciences, Bristol Medical School, University of Bristol, BristolBS2 8HW, UK
| | - Marco Fagnano
- Bristol Heart Institute, School of Translational Health Sciences, Bristol Medical School, University of Bristol, BristolBS2 8HW, UK
| | - Mohamed T Ghorbel
- Bristol Heart Institute, School of Translational Health Sciences, Bristol Medical School, University of Bristol, BristolBS2 8HW, UK
| |
Collapse
|
15
|
Jia Y, Li D, Yu J, Jiang W, Liu Y, Li F, Zeng R, Wan Z, Liao X. Angiogenesis in Aortic Aneurysm and Dissection: A Literature Review. Rev Cardiovasc Med 2023; 24:223. [PMID: 39076698 PMCID: PMC11266809 DOI: 10.31083/j.rcm2408223] [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: 01/04/2023] [Revised: 02/17/2023] [Accepted: 03/06/2023] [Indexed: 07/31/2024] Open
Abstract
Aortic aneurysm and aortic dissection (AA/AD) are critical aortic diseases with a hidden onset and sudden rupture, usually resulting in an inevitable death. Several pro- and anti-angiogenic factors that induce new capillary formation in the existing blood vessels regulate angiogenesis. In addition, aortic disease mainly manifests as the proliferation and migration of endothelial cells of the adventitia vasa vasorum. An increasing number of studies have shown that angiogenesis is a characteristic change that may promote AA/AD occurrence, progression, and rupture. Furthermore, neocapillaries are leaky and highly susceptible to injury by cytotoxic agents, which promote extracellular matrix remodeling, facilitate inflammatory cell infiltration, and release coagulation factors and proteases within the wall. Mechanistically, inflammation, hypoxia, and angiogenic factor signaling play important roles in angiogenesis in AA/AD under the complex interaction of multiple cell types, such as smooth muscle cells, fibroblasts, macrophages, mast cells, and neutrophils. Therefore, based on current evidence, this review aims to discuss the manifestation, pathological role, and underlying mechanisms of angiogenesis involved in AA/AD, providing insights into the prevention and treatment of AA/AD.
Collapse
Affiliation(s)
- Yu Jia
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Dongze Li
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China Hospital, Sichuan University West China School of Medicine, 610044 Chengdu, Sichuan, China
| | - Jing Yu
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China Hospital, Sichuan University West China School of Medicine, 610044 Chengdu, Sichuan, China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Yi Liu
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China Hospital, Sichuan University West China School of Medicine, 610044 Chengdu, Sichuan, China
| | - Fanghui Li
- Department of Cardiology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Rui Zeng
- Department of Cardiology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Zhi Wan
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China Hospital, Sichuan University West China School of Medicine, 610044 Chengdu, Sichuan, China
| | - Xiaoyang Liao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| |
Collapse
|
16
|
Gao J, Li L, Zhou D, Sun X, Cui L, Yang D, Wang X, Du P, Yuan W. Effects of norepinephrine‑induced activation of rat vascular adventitial fibroblasts on proliferation and migration of BMSCs involved in vascular remodeling. Exp Ther Med 2023; 25:290. [PMID: 37206559 PMCID: PMC10189611 DOI: 10.3892/etm.2023.11989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/11/2023] [Indexed: 05/21/2023] Open
Abstract
Vascular remodeling caused by vascular injury such as hypertension and atherosclerosis is a complex process involving a variety of cells and factors, and the mechanism is unclear. A vascular injury model was simulated by adding norepinephrine (NE) to culture medium of vascular adventitial fibroblasts (AFs). NE induced activation and proliferation of AFs. To investigate the association between the AFs activation and bone marrow mesenchymal stem cells (BMSCs) differentiation in vascular remodeling. BMSCs were cultured with supernatant of the AFs culture medium. BMSC differentiation and migration were observed by immunostaining and Transwell assay, respectively, while cell proliferation was measured using the Cell Counting Kit-8. Expression levels of smooth muscle actin (α-SMA), TGF-β1 and SMAD3 were measured using western blot assay. The results indicated that compared with those in the control group, in which BMSCs were cultured in normal medium, expression levels of α-SMA, TGF-β1 and SMAD3 in BMSCs cultured in medium supplemented with supernatant of AFs, increased significantly (all P<0.05). Activated AFs induced the differentiation of BMSCs into vascular smooth muscle-like cells and promoted proliferation and migration. AFs activated by NE may induce BMSCs to participate in vascular remodeling. These findings may help design and develop new approaches and therapeutic strategies for vascular injury to prevent pathological remodeling.
Collapse
Affiliation(s)
- Jun Gao
- Medical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Li Li
- Pediatric Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Dongli Zhou
- Nurse's Office, Health School of Laiyang, Laiyang, Yantai, Shandong 265200, P.R. China
| | - Xuhong Sun
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Lilu Cui
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Donglin Yang
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xiaohui Wang
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Pengchao Du
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
- Correspondence to: Professor Wendan Yuan or Professor Pengchao Du, Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong 264003, P.R. China E-mail: 981713509 @qq.com
| | - Wendan Yuan
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
- Correspondence to: Professor Wendan Yuan or Professor Pengchao Du, Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong 264003, P.R. China E-mail: 981713509 @qq.com
| |
Collapse
|
17
|
Wu J, Ma W, Qiu Z, Zhou Z. Roles and mechanism of IL-11 in vascular diseases. Front Cardiovasc Med 2023; 10:1171697. [PMID: 37304948 PMCID: PMC10250654 DOI: 10.3389/fcvm.2023.1171697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/28/2023] [Indexed: 06/13/2023] Open
Abstract
Vascular diseases are the leading cause of morbidity and mortality worldwide. Therefore, effective treatment strategies that can reduce the risk of vascular diseases are urgently needed. The relationship between Interleukin-11 (IL-11) and development of vascular diseases has gained increasing attention. IL-11, a target for therapeutic research, was initially thought to participate in stimulating platelet production. Additional research concluded that IL-11 is effective in treating several vascular diseases. However, the function and mechanism of IL-11 in these diseases remain unknown. This review summarizes IL-11 expression, function, and signal transduction mechanism. This study also focuses on the role of IL-11 in coronary artery disease, hypertension, pulmonary hypertension, cerebrovascular disease, aortic disease, and other vascular diseases and its potential as a therapeutic target. Consequently, this study provides new insight into the clinical diagnosis and treatment of vascular diseases.
Collapse
Affiliation(s)
- Jiacheng Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenrui Ma
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Wuhan, China
| | - Zhihua Qiu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihua Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Pisano C, Terriaca S, Scioli MG, Nardi P, Altieri C, Orlandi A, Ruvolo G, Balistreri CR. The Endothelial Transcription Factor ERG Mediates a Differential Role in the Aneurysmatic Ascending Aorta with Bicuspid or Tricuspid Aorta Valve: A Preliminary Study. Int J Mol Sci 2022; 23:10848. [PMID: 36142762 PMCID: PMC9502538 DOI: 10.3390/ijms231810848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
The pathobiology of ascending aorta aneurysms (AAA) onset and progression is not well understood and only partially characterized. AAA are also complicated in case of bicuspid aorta valve (BAV) anatomy. There is emerging evidence about the crucial role of endothelium-related pathways, which show in AAA an altered expression and function. Here, we examined the involvement of ERG-related pathways in the differential progression of disease in aortic tissues from patients having a BAV or tricuspid aorta valve (TAV) with or without AAA. Our findings identified ERG as a novel endothelial-specific regulator of TGF-β-SMAD, Notch, and NO pathways, by modulating a differential fibrotic or calcified AAA progression in BAV and TAV aortas. We provided evidence that calcification is correlated to different ERG expression (as gene and protein), which appears to be under control of Notch signaling. The latter, when increased, associated with an early calcification in aortas with BAV valve and aneurysmatic, was demonstrated to favor the progression versus severe complications, i.e., dissection or rupture. In TAV aneurysmatic aortas, ERG appeared to modulate fibrosis. Therefore, we proposed that ERG may represent a sensitive tissue biomarker to monitor AAA progression and a target to develop therapeutic strategies and influence surgical procedures.
Collapse
Affiliation(s)
- Calogera Pisano
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Sonia Terriaca
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Maria Giovanna Scioli
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Paolo Nardi
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Claudia Altieri
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Augusto Orlandi
- Pathological Anatomy, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
- Department of Biomedical Sciences, Catholic University of Our Lady of Good Counsel, 1001 Tirana, Albania
| | - Giovanni Ruvolo
- Department of Cardiac Surgery, Tor Vergata University Polyclinic, 00133 Rome, Italy
| | - Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy
| |
Collapse
|