1
|
Huang S, Liu D, Deng K, Shu C, Wu Y, Zhou Z. A computed tomography angiography-based radiomics model for prognostic prediction of endovascular abdominal aortic repair. Int J Cardiol 2025; 429:133138. [PMID: 40090490 DOI: 10.1016/j.ijcard.2025.133138] [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: 01/21/2025] [Revised: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
OBJECTIVE This study aims to develop a radiomics machine learning (ML) model that uses preoperative computed tomography angiography (CTA) data to predict the prognosis of endovascular aneurysm repair (EVAR) for abdominal aortic aneurysm (AAA) patients. METHODS In this retrospective study, 164 AAA patients underwent EVAR and were categorized into shrinkage (good prognosis) or stable (poor prognosis) groups based on post-EVAR sac regression. From preoperative AAA and perivascular adipose tissue (PVAT) image, radiomics features (RFs) were extracted for model creation. Patients were split into 80 % training and 20 % test sets. A support vector machine model was constructed for prediction. Accuracy is evaluated via the area under the receiver operating characteristic curve (AUC). RESULTS Demographics and comorbidities showed no significant differences between shrinkage and stable groups. The model containing 5 AAA RFs (which are original_firstorder_InterquartileRange, log-sigma-3-0-mm-3D_glrlm_GrayLevelNonUniformityNormalized, log-sigma-3-0-mm-3D_glrlm_RunPercentage, log-sigma-4-0-mm-3D_glrlm_ShortRunLowGrayLevelEmphasis, wavelet-LLH_glcm_SumEntropy) had AUCs of 0.86 (training) and 0.77 (test). The model containing 7 PVAT RFs (which are log-sigma-3-0-mm-3D_firstorder_InterquartileRange, log-sigma-3-0-mm-3D_glcm_Correlation, wavelet-LHL_firstorder_Energy, wavelet-LHL_firstorder_TotalEnergy, wavelet-LHH_firstorder_Mean, wavelet-LHH_glcm_Idmn, wavelet-LHH_glszm_GrayLevelNonUniformityNormalized) had AUCs of 0.76 (training) and 0.78 (test). Combining AAA and PVAT RFs yielded the highest accuracy: AUCs of 0.93 (training) and 0.87 (test). CONCLUSIONS Radiomics-based CTA model predicts aneurysm sac regression post-EVAR in AAA patients. PVAT RFs from preoperative CTA images were closely related to AAA prognosis after EVAR, enhancing accuracy when combined with AAA RFs. This preliminary study explores a predictive model designed to assist clinicians in optimizing therapeutic strategies during clinical decision-making processes.
Collapse
Affiliation(s)
- Shanya Huang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Dingxiao Liu
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Kai Deng
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chang Shu
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yan Wu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
2
|
Li X, Ma Y, Jiang Q, Zhan H, Sun X. The associations between circulating amino acids and arterial aneurysms and dissection: A bidirectional Mendelian randomization study. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2025; 25:200388. [PMID: 40160699 PMCID: PMC11951207 DOI: 10.1016/j.ijcrp.2025.200388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/13/2025] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
Background Circulating amino acid levels can be altered in arterial aneurysms and dissection, but the relationships between them is unclear. The present study investigated the causal relationship between circulating amino acid levels and arterial aneurysms and dissection via bidirectional two-sample Mendelian randomization (MR). Methods A bidirectional two-sample MR analysis was used. Forward analysis was performed with amino acid levels as the exposure and arterial aneurysms and dissection as outcomes. Reverse analysis was performed with arterial aneurysms and dissection as exposures and circulating amino acid levels as outcomes. MR data were analyzed using five analytical methods: the inverse-variance weighted (IVW), MR‒Egger, weighted median, simple, and weighted methods. IVW was used as the main analytical method, and the other methods were used for supplementary analyses. Heterogeneity was assessed using Cochran's Q test, and horizontal pleiotropy was assessed using intercepts from MR‒Egger regression. The genome-wide association study (GWAS) data for circulating amino acids were obtained from the IEU open GWAS database and the GWAS Catalog database. The GWAS data for arterial aneurysms and dissection were obtained from the Finngen consortium database version R10. Results The tyrosine level was negatively correlated with other aneurysms (P = 0.00211, OR: 0.57, 95 % CI: 0.40, 0.82). Aortic dissection decreased the circulating glycine level (P = 0.00168, OR: 0.98, 95 % CI: 0.98, 0.99). Conclusion Through bidirectional MR analysis, we found that tyrosine level was negatively correlated with other aneurysms and that aortic dissection reduced circulating glycine. Our findings support a possible interaction between circulating amino acid levels and arterial aneurysms and dissection.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yarong Ma
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qiulin Jiang
- Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Huizhi Zhan
- Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xiaolei Sun
- Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Laboratory of Nucleic Acids in Medicine for National High-level Talent, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, China
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Research Excellence, Faculty of Life Science and Medicine, King's College London, London, SE5 9NU, United Kingdom
| |
Collapse
|
3
|
Kopacz A, Kloska D, Bar A, Targosz-Korecka M, Cysewski D, Awsiuk K, Piechota-Polanczyk A, Cichon M, Chlopicki S, Jozkowicz A, Grochot-Przeczek A. Endothelial miR-34a deletion guards against aneurysm development despite endothelial dysfunction. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167812. [PMID: 40139409 DOI: 10.1016/j.bbadis.2025.167812] [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: 11/08/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
We previously reported a link between NRF2, a cytoprotective transcription factor, and the ageing of endothelial cells (ECs) and aorta. We also found that NRF2 KO mice are more susceptible to the development of abdominal aortic aneurysm (AAA), which is an age-associated condition. Since miR-34a is a marker of ageing, we explored its relationship with NRF2 and its role in vascular function and AAA formation. Here, we demonstrate that premature NRF2-dependent ageing of ECs is mediated by miR-34a. Infusion of hypertensive angiotensin II (Ang II) in mice increases miR-34a in the aortic endothelial layer and serum, particularly in mice developing AAA. Mice lacking endothelial miR-34a exhibit severe EC dysfunction. Despite that, they are protected from AAA, also on the NRF2 KO background. This protective effect is reversed by rapamycin, which suppresses Ang II-induced EC proliferation. We identified MTA2, but not SIRT1, as a target of miR-34a that inhibits EC proliferation stimulated by Ang II. These findings suggest that fine-tuning of EC proliferation could have potential therapeutic implications for the treatment of aneurysms.
Collapse
Affiliation(s)
- Aleksandra Kopacz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Damian Kloska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Marta Targosz-Korecka
- Department of Physics of Nanostructures and Nanotechnology, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Krakow, Poland
| | - Dominik Cysewski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Kamil Awsiuk
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Krakow, Poland
| | - Aleksandra Piechota-Polanczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Milena Cichon
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
4
|
Jia L, Wang Y, Jin C, Ma Y, Wang Y, Song L, Shen J, Xie Y, Xiang M. Macrophage heme oxygenase-1 modulates peroxynitrite-mediated vascular injury and exacerbates abdominal aortic aneurysm development. Am J Physiol Cell Physiol 2025; 328:C1808-C1821. [PMID: 40261277 DOI: 10.1152/ajpcell.00525.2023] [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: 10/11/2023] [Revised: 11/01/2023] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
Inflammatory reactions mediated by macrophages are profoundly related to the depletion of smooth muscle cells (SMCs) in abdominal aortic aneurysm (AAA) development. The findings from our previous investigation indicate that heme oxygenase-1 (HO-1) in macrophages exacerbates proinflammatory responses and oxidative damage. Therefore, the aim of this work was to gain insight into the function of HO-1 derived from macrophages and elucidate the underlying molecular mechanisms involved in AAA development. In this study, we discovered a dramatic increase in HO-1 expression in the infiltrated macrophages in experimental calcium phosphate-induced AAA tissues. Myeloid conditional HO-1-deficient mice displayed slower luminal area enlargement, as well as diminished inducible nitric oxide synthase (iNOS)-positive M1 macrophage activation, peroxynitrite generation, and SMCs apoptosis in aneurysmal tissues compared with littermate controls. Furthermore, we showed that inhibiting HO-1 eliminated the protein expression of iNOS induced by lipopolysaccharide/interferon-γ in bone marrow-derived macrophages, whereas the mRNA expression remained unaffected. Suppressing iNOS in macrophages alleviated SMCs apoptosis by decreasing nitric oxide generation in a coculture system in vitro. In summary, our study illustrates that macrophage-derived HO-1 strengthens AAA development through boosting the production of iNOS-dependent peroxynitrite and the deterioration of SMCs. These findings reveal potential therapeutic targets for resolving aneurysmal diseases.NEW & NOTEWORTHY This article illustrates the role of macrophage-derived heme oxygenase-1 (HO-1) in the development of abdominal aortic aneurysm (AAA). HO-1 deletion in macrophages hindered AAA development by reducing inducible nitric oxide synthase (iNOS)-dependent peroxynitrite production and smooth muscle cells (SMCs) apoptosis in vivo. Mechanistically, inhibition of HO-1 reduced the stimulated iNOS protein production in macrophages by lipopolysaccharide/interferon-γ. Moreover, suppressing iNOS in macrophages prevented SMCs apoptosis by decreasing nitric oxide generation in vitro.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/enzymology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Macrophages/enzymology
- Macrophages/pathology
- Peroxynitrous Acid/metabolism
- Nitric Oxide Synthase Type II/metabolism
- Nitric Oxide Synthase Type II/genetics
- Apoptosis
- Mice
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Heme Oxygenase-1/metabolism
- Heme Oxygenase-1/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Male
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Disease Models, Animal
- Aorta, Abdominal/pathology
- Aorta, Abdominal/enzymology
- Nitric Oxide/metabolism
- Membrane Proteins
Collapse
Affiliation(s)
- Liangliang Jia
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yufei Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Chunna Jin
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yuankun Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yidong Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Liuguang Song
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jian Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
5
|
Wu W, Tang W, Liang W, Li Q, Qi X, Gao R, Zhou P, Chen G, Xiong J, Gu X, Ding P, Yu M, Yang C, Wen S. GDF15 suppresses abdominal aortic aneurysm by upregulating AREG expression to adjust macrophage polarization. Int Immunopharmacol 2025; 159:114899. [PMID: 40414071 DOI: 10.1016/j.intimp.2025.114899] [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: 10/22/2024] [Revised: 01/19/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVE Inflammation plays a key role in abdominal aortic aneurysm (AAA), with macrophages being crucial. Growth differentiation factor 15 (GDF15) is a new anti-inflammatory cytokine potentially useful in AAA diagnosis and treatment, but its role is unclear. METHODS AND RESULTS In mice with AAA, GDF15 expression was higher in lesioned tissues. Daily intraperitoneal injection of recombinant GDF15 (rGDF15) reduced aortic dilation, inflammation, degradation of aortic wall elastin and collagen, cellular apoptosis, and increased smooth muscle cells. GDF15 knockdown worsened AAA severity. Immunohistochemistry and immunofluorescence showed rGDF15 treatment reduced M1 macrophage polarization and enhanced M2 polarization, decreasing the M1/M2 ratio. GDF15 knockdown had the opposite effect. Additionally, Amphiregulin (AREG) expression increased with rGDF15 treatment and decreased with GDF15 knockdown. Immunofluorescence colocalization revealed lower AREG expression in M1 macrophages and higher AREG expression in M2 macrophages, suggesting that AREG may be involved in the regulation of macrophage polarization by GDF15 in AAA. Mechanistically, GDF15 upregulates AREG expression by activating the TGF-βR/SMAD2/3 signaling pathway, thereby inhibiting M1 polarization and promoting M2 polarization of macrophages. CONCLUSION This study demonstrates that exogenous injection of rGDF15 upregulates AREG expression and regulates macrophage polarization, thereby inhibiting AAA. GDF15 may not only serve as a diagnostic and prognostic marker for AAA but also as a potential molecular target for therapeutic intervention in AAA.
Collapse
Affiliation(s)
- Wanying Wu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenjing Tang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Liang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoyu Qi
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ran Gao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gezheng Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinrong Xiong
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoying Gu
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peiwu Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Miao Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chao Yang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Shuang Wen
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
6
|
Yang WT, Li FD, Zheng YH, Wang L. Myeloid Cells in Abdominal Aortic Aneurysm. Curr Atheroscler Rep 2025; 27:57. [PMID: 40402405 DOI: 10.1007/s11883-025-01302-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE OF REVIEW Abdominal aortic aneurysm (AAA) is a life-threatening vascular disorder with high mortality upon rupture, yet effective pharmacotherapy remains lacking. This review synthesizes the pivotal roles of myeloid cells-key mediators of aortic inflammation and remodeling-in AAA pathogenesis, highlighting their therapeutic targeting potential. RECENT FINDINGS Single-cell RNA sequencing has revealed myeloid diversity in AAA. Among these myeloid populations, macrophages (including interferon-responsive monocytes, pro- and anti-inflammatory subsets, and reparative populations) emerge as central regulators of AAA pathogenesis, influencing disease initiation, progression, and tissue repair processes. Neutrophils promote vascular injury via neutrophil extracellular traps, while dendritic cells bridge innate-adaptive immunity. Eosinophils and myeloid-derived suppressor cells exhibited protective effects by immunoregulation. Mechanistic studies identified transcriptional, metabolic, and epigenetic regulators of myeloid plasticity. Clonal hematopoiesis and trained immunity may serve as potential novel mechanisms of myeloid cells involved in AAA. These mechanistic insights have inspired therapeutic innovation, with nanoparticle-targeted myeloid cell therapies showing promising immunomodulatory effects in mitigating AAA progression. Myeloid cells play a pivotal role in AAA pathogenesis by driving inflammatory responses, extracellular matrix degradation, and maladaptive vascular remodeling. Their functional heterogeneity, encompassing both destructive and protective subsets, highlights the need for precisely targeted therapeutic approaches. While single-cell technologies have significantly advanced our understanding of myeloid diversity, clinical translation remains challenged by microenvironmental crosstalk and potential off-target effects. Future research should prioritize: (1) spatial multi-omics characterization of myeloid-vascular interactions, (2) development of precision therapies targeting clonal hematopoiesis-driven subpopulations, and (3) combinatorial strategies to reprogram pathogenic myeloid phenotypes. Addressing these critical gaps may lead to transformative therapies for aneurysm stabilization, ultimately fulfilling the urgent unmet needs in AAA clinical management.
Collapse
Affiliation(s)
- Wen-Tao Yang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Dongcheng District, 100730, Beijing, China
| | - Fang-Da Li
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Dongcheng District, 100730, Beijing, China
| | - Yue-Hong Zheng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Dongcheng District, 100730, Beijing, China.
| | - Lei Wang
- Department of Nutrition and Food Hygiene, Beijing Key Laboratory of Environment and Aging, School of Public Health, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
7
|
Pan X, Zhang R, Lu B, Chen S, Chen H, Li M, Qin L, Song Z, Yang Y, Wang Z, Yan F, Sun A, Wu F, Zhang L, Wang J, Guzik TJ, Gao P. SM22α-Lineage Perivascular Stromal Cells Contribute to Abdominal Aortic Aneurysm. Circ Res 2025. [PMID: 40371535 DOI: 10.1161/circresaha.124.325750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Perivascular adipose tissue (PVAT) is a key regulator of vascular dysfunction. Impairment of PVAT phenotypic plasticity with aging may play a role in vascular pathology including abdominal aortic aneurysms (AAAs). Yet, the mechanisms underlying PVAT plasticity in aneurysm pathogenesis remain elusive. METHODS Single-cell RNA sequencing was performed on perivascular stromal cells from young (2- to 3-month-old) and aged (18- to 20-month-old) mice. The expression of PGC-1α (peroxisome proliferator-activated receptor gamma coactivator-1α) was measured in PVAT of aged mice and human aneurysm samples. Loss- and gain-of-function approaches were used to investigate the role of SM22α-lineage perivascular stromal cell-derived PGC-1α in aneurysm development. Molecular mechanisms were explored through transcriptome and functional studies in young and aged mice, SM22αCre; Rosa26RFP/+; PGC1αf/f and SM22αCre; Rosa26RFP/+ mice with Ang II (angiotensin II)-induced and deoxycorticosterone acetate/salt-induced AAA models. RESULTS SM22α+ cells accumulated in PVAT of Ang II-treated aged mice and patients with aortic aneurysms. Single-cell RNA sequencing analysis revealed that aging disrupted the differentiation potential of SM22α-lineage perivascular stromal cells and led to reduced PGC-1α levels. PGC1α downregulation in PVAT was observed in both mouse AAA models and human aneurysm lesions. In mice with SM22α-driven PGC-1α deletion, Ang II-induced AAA formation was accompanied by perivascular stromal cell-to-myofibroblast differentiation. In vitro PGC1α knockdown suppressed nuclear YAP (Yes-associated protein) signaling, reducing adipocyte differentiation, while increasing MMP2 (matrix metalloproteinase 2)-secreting myofibroblasts. Furthermore, PGC-1α overexpression in aged mice or administration of the YAP signaling inhibitor verteporfin in SM22αCre; Rosa26RFP/+; PGC1αf/f mice restored PVAT function and conferred protection against aneurysm formation. Last, we used the radiomics analysis to noninvasively evaluate PVAT in the context of AAA severity in humans. CONCLUSIONS PGC-1α deficiency in SM22α-lineage stromal cells disrupts the balance between adipogenic and myofibrogenic differentiation through regulating YAP signaling, ultimately promoting aneurysm development. Radiomics assessment may present a promising noninvasive approach for PVAT evaluation in aneurysms, offering valuable potential for clinical research.
Collapse
Affiliation(s)
- Xiaoxi Pan
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Run Zhang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Bingling Lu
- School of Biomedical Engineering, Shanghai Jiao Tong University, China (B.L., Z.S., L.Z.)
| | - Siyuan Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Hongjin Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Mengyao Li
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Le Qin
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (L.Q., F.Y.)
| | - Zhiyun Song
- School of Biomedical Engineering, Shanghai Jiao Tong University, China (B.L., Z.S., L.Z.)
| | - Yi Yang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (Y.Y., Z.W.)
| | - Zhe Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (Y.Y., Z.W.)
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (L.Q., F.Y.)
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China (A.S.)
| | - Fang Wu
- Shanghai Clinical Research Center for Aging and Medicine, Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (F.W.)
| | - Lichi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, China (B.L., Z.S., L.Z.)
| | - Jiguang Wang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Tomasz J Guzik
- Department of Medicine, Jagiellonian University, Colelgium Medicum, Krakow, Poland (T.J.G.)
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, United Kingdom (T.J.G.)
| | - Pingjin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| |
Collapse
|
8
|
Tan Y, Zhu X, Huang Y, Zhao C, Cheng X, Li J, Zhang G, Ma T, Yang S, Bai Y. Clonal Hematopoiesis of Indeterminate Potential Is Associated With Incident Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2025. [PMID: 40336477 DOI: 10.1161/atvbaha.124.322630] [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: 02/14/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP) is an emerging risk factor for cardiovascular diseases. Genetic IL (interleukin)-6 signaling deficiency reduced cardiovascular disease risk in CHIP carriers. However, the association between CHIP and incident abdominal aortic aneurysm (AAA) and whether IL-6 signaling inhibition attenuates AAA risk among individuals with CHIP remained unclear. METHODS Participants without prevalent AAA from the UK Biobank were included. The associations of any CHIP (variant allele fraction, ≥2%), large CHIP (variant allele fraction, ≥10%), and gene-specific CHIP subtypes with incident AAA were investigated. The protection role of IL6R p.Asp358Ala, a genetic proxy for IL-6 deficiency, was tested after stratification by CHIP status. Furthermore, the interaction and joint effects of CHIP and genetic susceptibility on AAA risk were tested. RESULTS This study included 425 211 participants. Any CHIP and large CHIP was identified in 13 768 (3.2%) and 8576 (2.0%) participants, respectively. CHIP was associated with an increased risk of incident AAA (hazard ratio [HR], 1.21 [95% CI, 1.01-1.44]; P=0.034), with large CHIP clones exhibiting greater effect size (HR, 1.35 [95% CI, 1.10-1.66]; P=0.0045). Driver gene-specific analyses revealed that ASXL1-mediated CHIP exerted the strongest effect size on AAA risk (HR, 2.10 [95% CI, 1.54-2.88]; P<0.001). The presence of 2 IL6R p.Asp358Ala alleles attenuated the risk of AAA in large CHIP carriers (HR, 0.48 [95% CI, 0.23-0.99]; P=0.046). In the joint analysis, participants with CHIP and high genetic risk had a higher risk of developing AAA than those without CHIP and with low genetic risk (HR, 2.15 [95% CI, 1.63-2.85]; P<0.001). CONCLUSIONS CHIP is associated with an increased risk of AAA. Genetic IL-6 signaling deficiency attenuates the risk of AAA in large CHIP carriers. CHIP may serve as an attractive target for the prevention and treatment of AAA.
Collapse
Affiliation(s)
- Yu Tan
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Xuanmeng Zhu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Yuanfeng Huang
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. (Y.H., J.L.)
- Bioinformatics Center, Xiangya Hospital and Furong Laboratory, Central South University, Changsha, Hunan, China. (Y.H., J.L.)
| | - Chenxuan Zhao
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Xunjie Cheng
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Jinchen Li
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. (Y.H., J.L.)
- Bioinformatics Center, Xiangya Hospital and Furong Laboratory, Central South University, Changsha, Hunan, China. (Y.H., J.L.)
| | - Guogang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Tianqi Ma
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Shujun Yang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Yongping Bai
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.B.)
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.B.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.B.)
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China. (Y.B.)
| |
Collapse
|
9
|
Gautier EL. Endothelium calls macrophages for destruction. Nat Immunol 2025; 26:644-646. [PMID: 40263615 DOI: 10.1038/s41590-025-02123-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Affiliation(s)
- Emmanuel L Gautier
- Sorbonne Université, Inserm, Nutrition et obésités: approches systémiques, Paris, France.
| |
Collapse
|
10
|
Liu X, Li Z, Xu H, He W, Wu L, Ji B, Nuermaimaiti N, Ao G, Feng Y, He X. Revealing shared molecular and mechanistic signatures between intracranial aneurysms and abdominal aortic aneurysms: a comprehensive genomic analysis. Orphanet J Rare Dis 2025; 20:196. [PMID: 40275351 PMCID: PMC12020248 DOI: 10.1186/s13023-025-03689-1] [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: 04/22/2024] [Accepted: 03/19/2025] [Indexed: 04/26/2025] Open
Abstract
Intracranial aneurysms (IAs) and abdominal aortic aneurysms (AAAs) are both vascular diseases that are closely linked. However, the pathogenesis underlying the co-occurrence of IAs and AAAs remains poorly understood. This study aims to identify key biomarkers that shed light on the molecular mechanisms connecting these two diseases using bioinformatics analysis. Gene expression profiles (GSE122897, GSE237229) were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) common to both IAs and AAAs were identified and subjected to functional enrichment analysis. The Cytoscape cytoHubba plugin was used to identify hub genes, and their predictive ability was evaluated using the receiver operating characteristic (ROC) curve. Additionally, immune infiltration analyses and single-gene gene set enrichment analysis (GSEA) were conducted for the hub genes. A total of 46 DEGs were identified, including 40 upregulated genes and 6 downregulated genes. The common DEGs were found to be involved in extracellular matrix structural constituents, collagen fibril organization, and regulation of basic cellular processes. ITGA11 was identified as a key gene implicated in the comorbidity of IAs and AAAs, with its upregulation strongly associated with plasma cells. Furthermore, in both IAs and AAAs, glycosaminoglycan biosynthesis of extracellular matrix components and immune-related diseases were significantly linked to the high expression of ITGA11. Our findings suggest that the comorbidity of IAs and AAAs may be driven by shared inflammatory and immune response mechanisms, with ITGA11 emerging as a potential biomarker for this co-occurrence.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, Guangzhou, 510282, China
- Department of Neurosurgery, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Zhenjun Li
- Brain Vascular Disease Center, Guangdong Second Provincial People's Hospital, Guangzhou, 510317, China
| | - Hongzhen Xu
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Wangqing He
- Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, Guangzhou, 510282, China
| | - Lei Wu
- Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, Guangzhou, 510282, China
| | - Bin Ji
- Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, Guangzhou, 510282, China
| | - Nuerzhati Nuermaimaiti
- Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, Guangzhou, 510282, China
| | - Guangnan Ao
- Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, Guangzhou, 510282, China
| | - Yuhang Feng
- Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, Guangzhou, 510282, China
| | - Xuying He
- Brain Vascular Disease Center, Guangdong Second Provincial People's Hospital, Guangzhou, 510317, China.
| |
Collapse
|
11
|
Tang Y, Guo T, Wang X, Li C, Zhang X, Zhang J. Cyclodextrin-Derived Macromolecular Therapies for Inflammatory Diseases. Macromol Biosci 2025:e2400637. [PMID: 40271896 DOI: 10.1002/mabi.202400637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Inflammation is an essential physiological defense mechanism against harmful stimuli, yet dysregulated inflammatory responses are closely associated with the pathogenesis of numerous acute and chronic diseases. Recent advances highlight the remarkable anti-inflammatory potential of bioactive macromolecules, particularly cyclodextrins (CDs) and their engineered derivatives, which are emerging as promising therapeutic agents. This review systematically introduces different CDs and CD-derived macromolecules that demonstrate anti-inflammatory properties, with emphasis on their molecular mechanisms of action. Native CDs exhibit direct therapeutic effects through host-guest interactions, enabling selective sequestration of pathogenic components such as cholesterol crystals and proteins that drive inflammatory cascades. Moreover, chemically modified CD derivatives incorporating functional groups demonstrate enhanced capabilities in neutralizing inflammatory mediators and modulating immune cell responses. This work further discusses the expanding therapeutic applications of these macromolecules across diverse inflammatory conditions, ranging from acute tissue injuries to chronic autoimmune disorders. Finally, this work critically analyzes the crucial challenges and emerging opportunities in translating CD-based macromolecular therapies into clinical practice, addressing key considerations in biocompatibility, targeted delivery, and therapeutic efficacy optimization.
Collapse
Affiliation(s)
- Yige Tang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- International Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Tao Guo
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xuanran Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiangjun Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Yu-Yue Pathology Scientific Research Center, 313 Gaoteng Avenue, Jiulongpo District, Chongqing, 400039, China
| |
Collapse
|
12
|
Erdemutu E, Zhou C, Ma M, Hu L, Wu J, Dai X, Gao Z. Endovascular repair of abdominal aortic aneurysm-related type II endoleak: a multicenter study on the possibility of further intervention. Front Cardiovasc Med 2025; 12:1450942. [PMID: 40313581 PMCID: PMC12043675 DOI: 10.3389/fcvm.2025.1450942] [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: 06/18/2024] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Background We aimed to analyze the risk factors associated with Type II endoleak (T2EL) requiring reintervention after endovascular aneurysm repair (EVAR) for multicenter abdominal aortic aneurysms. Methods A retrospective analysis was conducted on data from 614 patients with abdominal aortic aneurysms who underwent elective EVAR at three centers (Tianjin Medical University General Hospital, Affiliated Hospital of Inner Mongolia Medical University, Shanxi Provincial People's Hospital) from January 2017 to December 2021. After applying exclusion criteria, 375 patients were included in the study, with 50 patients in the T2EL-related reintervention group and 325 patients in the non-T2EL group. Single-factor and multiple-factor logistic analyses were used to identify high-risk factors, and ROC curve analysis was performed to determine the risk thresholds for mesenteric artery diameter, number of lumbar arteries, maximum aneurysm diameter, and proportion of intraluminal thrombus volume. Results The rate of T2EL-related reintervention among the 375 patients was 13.33% (50/375). Single-factor analysis indicated that age, hypertension, maximum aneurysm diameter, proportion of intraluminal thrombus, diameter of inferior mesenteric artery (IMA), and number of patent lumbar arteries (LA) were risk factors for T2EL-related reintervention. Multiple-factor logistic analysis identified maximum aneurysm diameter, proportion of thrombus, IMA diameter, and number of patent LA as the main influencing factors for T2EL-related reintervention after EVAR. Significant risk factors for reintervention were maximum aneurysm diameter (OR = 1.043, 95% CI 1.015-1.072, P = 0.002), IMA diameter (OR = 3.901, 95% CI 1.116-13.632, P = 0.033), and number of LA (OR = 2.584, 95% CI 1.722-3.769, P < 0.001). A significant protective factor for reintervention was thrombus proportion (OR = 0.895, 95% CI 0.864-0.927, P < 0.001). ROC curve analysis showed that the risk thresholds for reintervention were an IMA diameter of 2.95 mm, intraluminal thrombus volume proportion <42.5%, number of LA ≤5.5, and aneurysm diameter of 53.55 mm. Conclusion Cases with identified risk factors are considered to have a higher risk of T2EL-related reintervention after EVAR. Exceeding the new risk thresholds may indicate a higher likelihood of T2EL-related reintervention after EVAR.
Collapse
Affiliation(s)
- E. Erdemutu
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Department of Vascular Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Chongbin Zhou
- Department of Vascular Surgery, Hohhot First Hospital, Hohhot, China
| | - Ming Ma
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Department of Vascular Surgery, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Liqiang Hu
- Department of Vascular Surgery, Hohhot First Hospital, Hohhot, China
| | - Jisiguleng Wu
- Department of Vascular Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xiangchen Dai
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhanfeng Gao
- Department of Vascular Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
13
|
Bi C, Liu B, Gao P, Wang C, Fang S, Huo Z, Song Q, Dong D, Wu X, Li G. RAGE deficiency ameliorates abdominal aortic aneurysm progression. Inflamm Res 2025; 74:63. [PMID: 40244438 DOI: 10.1007/s00011-025-02027-2] [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: 12/15/2024] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a vascular disease characterized by inflammation and arterial wall degradation. The receptor for advanced glycation end products (RAGE) plays a pivotal role in regulating inflammatory pathways, but its specific contribution to AAA pathogenesis remains unclear. PURPOSE This study aimed to investigate the role of RAGE in AAA development by examining its expression in human and mice AAA tissues and exploring the effects of RAGE deficiency on aneurysm progression, macrophage polarization, and inflammatory responses. METHODS RAGE expression was analyzed in human AAA samples and porcine pancreatic elastase (PPE) induced AAA mouse models using Western blotting, immunohistochemistry, and immunofluorescence. In vivo RAGE-deficient (RAGE-/-) mice were generated to assess the impact of RAGE knockout on AAA progression. In vitro experiments utilized RAW264.7 transfected with RAGE-targeting siRNA to study macrophage polarization and NF-κB signaling. RESULTS RAGE expression was elevated in AAA tissues, particularly in macrophages. RAGE-/- mice exhibited reduced AAA incidence, mortality, and aortic dilation compared to wild-type mice. Histological analysis showed preserved elastic fibers and smooth muscle layers, along with decreased inflammatory cell infiltration and MMP2/MMP9 expression. RAGE deficiency inhibited M1-like macrophage polarization and pro-inflammatory cytokine secretion, mediated through suppression of the NF-κB pathway. CONCLUSIONS RAGE deficiency mitigates AAA progression by modulating macrophage polarization and reducing inflammation via the NF-κB pathway. These findings highlight RAGE as a potential therapeutic target for AAA treatment.
Collapse
Affiliation(s)
- Cong Bi
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Bingqi Liu
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Peixian Gao
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Chuanle Wang
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Sheng Fang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200030, China
| | - Zhengkun Huo
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Qingpeng Song
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Dianning Dong
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xuejun Wu
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Gang Li
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
14
|
Xu S, Han X, Yu Y, Qu C, Yang B, Shen B, Liu X. Deficiency of IL-7R attenuates abdominal aortic aneurysms in mice by inhibiting macrophage polarization towards M1 phenotype through the NF-κB pathway. Mol Med 2025; 31:138. [PMID: 40240976 PMCID: PMC12004661 DOI: 10.1186/s10020-025-01209-2] [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: 11/04/2024] [Accepted: 04/10/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a common degenerative disease of the abdominal aorta, which can result in extremely high mortality owing to the rupture of the abdominal aorta. The activation of IL-7R has been shown to modulate the inflammatory responses, which play an important role in the progression of AAAs. However, the mechanism of IL-7/IL-7R axis in AAAs is still unclear. AIMS This study aims to investigate the effects of IL-7R on AAAs and the underlying mechanisms involved. METHODS Wild-type C57BL/6 and IL-7R knockout mice were used as experimental subjects. ELISA analysis, histological staining, western blotting and qPCR were performed to explore effects of IL-7R deficiency in the formation and development of elastase-induced AAAs. Transwell, CCK8, and immunofluorescence assays detected the migration and polarization of RAW264.7 macrophages in vitro. RESULT We demonstrated that IL-7R was elevated in mice with AAAs. Blocking IL-7R can inhibit the formation of AAAs and reduce aortic dilatation, elastic layer degradation, and inflammatory cell infiltration. Knockout of IL-7R suppressed the migration, infiltration and M1 polarization of macrophages. Moreover, inhibition of the NF-κB signaling pathway by BAY 11-7082 attenuated the macrophage-mediated inflammatory responses caused by IL-7R overexpression. CONCLUSION In short, this study showed that IL-7R promotes the infiltration and migration of macrophages by regulating M1 macrophage polarization, possibly in part via activation of the NF-κB pathway, which may be associated with the development of AAAs.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/etiology
- Aortic Aneurysm, Abdominal/genetics
- NF-kappa B/metabolism
- Mice
- Macrophages/metabolism
- Macrophages/immunology
- Signal Transduction
- Mice, Knockout
- Disease Models, Animal
- Receptors, Interleukin-7/genetics
- Receptors, Interleukin-7/metabolism
- Receptors, Interleukin-7/deficiency
- Male
- Mice, Inbred C57BL
- RAW 264.7 Cells
- Phenotype
- Macrophage Activation
Collapse
Affiliation(s)
- Shengnan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, P.R. China
| | - Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, P.R. China
| | - Yi Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, P.R. China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, P.R. China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P.R. China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, P.R. China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, P.R. China.
| | - Bo Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P.R. China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, P.R. China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, P.R. China.
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P.R. China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, P.R. China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, P.R. China.
| |
Collapse
|
15
|
Xu H, Yang J, Wei Z, Bao S, Liu Z. Oxidative stress in vascular surgical diseases: mechanisms, impacts and therapeutic perspectives. Front Pharmacol 2025; 16:1527684. [PMID: 40271068 PMCID: PMC12014636 DOI: 10.3389/fphar.2025.1527684] [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: 11/13/2024] [Accepted: 03/24/2025] [Indexed: 04/25/2025] Open
Abstract
The role of oxidative stress in vascular surgical diseases has increasingly been recognized as significant. This paper systematically reviews the specific mechanisms of oxidative stress in a various vascular surgical condition, including aortic dissection, abdominal aortic aneurysm, thrombosis, diabetic foot, and thromboangiitis obliterans, while also exploring related therapeutic strategies. Oxidative stress arises from an imbalance between free radicals and antioxidants, where excess reactive oxygen species and other free radicals can exacerbate inflammatory response. This paper delves into the pathogenic mechanisms of oxidative stress in the aforementioned diseases and discusses potential methods for utilizing antioxidants to reduce oxidative stress levels. Additionally, this paper highlights the challenges faced by current antioxidant therapies and identifies future research directions. By summarizing current research progress, this paper aims to provide a theoretical basis for more effective treatment strategies of vascular surgical diseases, with the hope of advancing the field.
Collapse
Affiliation(s)
- Haosen Xu
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Jin Yang
- College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zhanhui Wei
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Shijie Bao
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Zhuo Liu
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
16
|
Jamalinia M, Lonardo A, Weiskirchen R. Abdominal Aortic Aneurysm and Liver Fibrosis: Clinical Evidence and Molecular Pathomechanisms. Int J Mol Sci 2025; 26:3440. [PMID: 40244390 PMCID: PMC11989544 DOI: 10.3390/ijms26073440] [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: 03/10/2025] [Revised: 03/29/2025] [Accepted: 04/05/2025] [Indexed: 04/18/2025] Open
Abstract
To stimulate further research, this review summarizes studies linking liver fibrosis with the risk of abdominal aortic aneurysms (AAA). AAA is defined as a permanently weakened and dilated abdominal aorta, which develops due to inflammation of the tunica media, activation of the renin-angiotensin-aldosterone system, immune system activation, and coagulation disorders. Typically asymptomatic, AAA is often incidentally detected through imaging done for abdominal symptoms or as part of screening programs. AAA follows a variable course and has a mortality rate strongly dependent on age and sex. Risk factors for AAA include age, male sex, ethnicity, family history of AAA, lifestyle habits, arterial hypertension, dyslipidemia, and comorbid atherosclerotic cardiovascular disease. Conversely, individuals with type 2 diabetes, female sex, and certain ethnicities are at a reduced risk of AAA. Liver fibrosis, resulting from chronic liver diseases owing to varying etiologies, is increasingly recognized as a potential contributor to AAA development. Evidence increasingly indicates that metabolic dysfunction-associated steatotic liver disease (MASLD) and other chronic liver conditions may intensify inflammatory pathways shared with AAA, thereby potentially exacerbating AAA progression. This review specifically examines the epidemiology and risk factors associated with the link between AAA and liver fibrosis. It also highlights potential pathomechanisms, including systemic inflammation, oxidative stress, and extracellular matrix remodeling, which may contribute to both conditions. Although these findings underscore significant overlaps in risk profiles, additional research is needed to clarify whether type 2 diabetes, female sex, and certain ethnicities truly confer protection against AAA or if this association is influenced by other confounding variables. Ultimately, addressing these open questions will help guide targeted therapeutic interventions and the identification of novel biomarkers to predict disease progression.
Collapse
Affiliation(s)
- Mohamad Jamalinia
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz 41100, Iran;
| | - Amedeo Lonardo
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| |
Collapse
|
17
|
Wang L, Lin Y, Lin Z, Wu Q, Zhong G, Chen L. Causal relationship between mitochondrial proteins and risks of aortic aneurysms and aortic dissection: a Mendelian randomization study. J Cardiothorac Surg 2025; 20:181. [PMID: 40186305 PMCID: PMC11971758 DOI: 10.1186/s13019-025-03389-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 03/09/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Mitochondrial dysfunction may be linked to the development of aortic aneurysm (AA) and aortic dissection (AD). This study aimed to evaluate the potential associations between proteins related to mitochondrial function and the risks of AA/AD using Mendelian randomization (MR). METHODS Large-scale publicly available genome-wide association studies (GWAS) and FinnGen summary data were utilized for MR analysis. The causal relationship between mitochondrial proteins and AA/AD was assessed using inverse-variance weighted (IVW) as the primary method. Sensitivity analyses were conducted to detect heterogeneity and pleiotropy by Cochran's Q test, MR-Egger test, MR-PRESSO global test, and "leave-one-out" analysis. RESULTS There were potential causal relationships between several mitochondrial proteins and AA/AD. Specifically, the iron-sulfur cluster assembly enzyme ISCU (OR = 1.165, 95% CI: 1.051-1.291, P = 0.004) and NFU1 iron-sulfur cluster scaffold homolog (OR = 1.184, 95% CI: 1.056-1.329, P = 0.004) were identified as potential risk factors for AA; whereas the 39 S ribosomal protein L14 (OR = 0.868, 95% CI: 0.764-0.987, P = 0.031) was found to be a protective factor for AA. Furthermore, 39 S ribosomal protein L33 (OR = 1.134, 95% CI: 1.010-1.274, P = 0.033) and cytochrome C oxidase subunit 5B (OR = 1.330, 95% CI: 1.037-1.706, P = 0.025) were associated with increased risks of AD; whereas the 39 S ribosomal protein L52 (OR = 0.736, 95% CI: 0.550-0.984, P = 0.038) and mitochondrial ubiquitin ligase activator of NFKB 1 (OR = 0.806, 95% CI: 0.656-0.989, P = 0.039) were identified as potential protective factors for AD. Sensitivity analysis confirmed the stability of the results. CONCLUSIONS This study identified potential genetic associations between mitochondrial proteins and AA/AD. Targeting these mitochondrial proteins may help prevent the occurrence of AA/AD.
Collapse
Affiliation(s)
- Lei Wang
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, Fujian province, China
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, 350000, Fujian province, China
| | - Yuzuo Lin
- Union College of Clinical Medicine, Fujian Medical University Union Hospital, Fuzhou, 350000, Fujian province, China
| | - Ziyan Lin
- Union College of Clinical Medicine, Fujian Medical University Union Hospital, Fuzhou, 350000, Fujian province, China
| | - Qingtong Wu
- Union College of Clinical Medicine, Fujian Medical University Union Hospital, Fuzhou, 350000, Fujian province, China
| | - Guodong Zhong
- Department of Pathology, Fujian Province Second People's Hospital, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, Fujian province, China.
| | - Liangwan Chen
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, Fujian province, China.
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, 350000, Fujian province, China.
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350000, Fujian province, China.
| |
Collapse
|
18
|
Liu Y, Yu M, Wang H, Dorsey KH, Cheng Y, Zhao Y, Luo Y, Zhao G, Zhao Y, Lu H, Deng Y, Mu W, Liu H, Wu X, Wang Z, Zhang J, Chang L, Chen YE, Schwendman A, Guo Y. Restoring Vascular Smooth Muscle Cell Mitochondrial Function Attenuates Abdominal Aortic Aneurysm in Mice. Arterioscler Thromb Vasc Biol 2025; 45:523-540. [PMID: 39945066 PMCID: PMC12038981 DOI: 10.1161/atvbaha.124.321730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/24/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a complex vascular pathology without pharmaceutical interventions. This study aimed to evaluate whether restoring vascular smooth muscle cell (VSMC) mitochondrial function could prevent AAA development. METHODS Ang II (angiotensin II)-induced AAA was established in Ldlr-deficient mice, and the gene expression profiles in abdominal aortic tissues exhibiting varying degrees of severity were analyzed. Synthetic high-density lipoprotein (sHDL) formulated with Apoa1 mimetic peptide and phospholipids was evaluated for the protective effects on VSMC mitochondria. The therapeutic efficacy of sHDL was further investigated in Ang II-infusion and PPE (porcine pancreatic elastase)-induced AAA models. RESULTS VSMC mitochondrial damage intensified gradually during AAA development, which was confirmed in distinct AAA animal models and human tissues. sHDL accumulated in the aneurysmatic lesions and restored mitochondrial DNA levels and the expression of genes related to oxidative phosphorylation following Ang II infusion. In mouse primary VSMCs, sHDL maintained mitochondrial homeostasis by suppressing the upregulation of DRP1 (dynamin-related protein 1), a protein involved in mitochondrial fission, reducing the generation of reactive oxygen species, preventing the loss of mitochondrial membrane potential, and preserving mitochondrial respiratory capacity. Administration of sHDL decreased Ang II-induced AAA incidence (control versus treatment, 76% versus 40%; P<0.05) and maximum aortic diameters. The protective effects of sHDL were further validated in the PPE model, with reductions observed in maximum aortic diameters and aortic mitochondrial DNA loss. Post-Ang II infusion, administration of sHDL improved VSMC mitochondrial function and suppressed aneurysm growth in Apoe-deficient mice. Human AAA is characterized by mitochondrial dysfunction, and liver-derived HDL (high-density lipoprotein) components play a pivotal role in regulating gene expression in aortic tissues. CONCLUSIONS VSMC mitochondrial damage is a pivotal factor in the development of AAA. The utilization of sHDL nanoparticles represents a promising novel therapeutic approach for AAA, aimed at restoring VSMC mitochondrial function.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/prevention & control
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Disease Models, Animal
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Angiotensin II
- Humans
- Male
- Mice, Inbred C57BL
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aorta, Abdominal/drug effects
- Dynamins/metabolism
- Dynamins/genetics
- Cells, Cultured
- Mice, Knockout
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Mitochondrial Dynamics/drug effects
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/pathology
- Mitochondria, Muscle/drug effects
- Membrane Potential, Mitochondrial/drug effects
- Reactive Oxygen Species/metabolism
- Mice
- DNA, Mitochondrial/metabolism
- DNA, Mitochondrial/genetics
- Oxidative Phosphorylation
- Mitochondria/metabolism
- Mitochondria/drug effects
- Mitochondria/pathology
Collapse
Affiliation(s)
- Yaozhong Liu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Contributed equally
| | - Minzhi Yu
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
- Contributed equally
| | - Huilun Wang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kristen Hong Dorsey
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Yalun Cheng
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Ying Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Yonghong Luo
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Guizhen Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Yang Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Haocheng Lu
- Department of Pharmacology, Southern University of Science and Technology, Guangdong, China
| | - Yongjie Deng
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Wenjuan Mu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Hongyu Liu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Xiaokang Wu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Zhenguo Wang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Lin Chang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Y. Eugene Chen
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Anna Schwendman
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Yanhong Guo
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Zhang Q, Cai Z, Yu Z, Di C, Qiu Y, Qi R. Agathis dammara Extract and its Monomer Araucarone Attenuate Abdominal Aortic Aneurysm in Mice. Cardiovasc Drugs Ther 2025; 39:239-257. [PMID: 37979015 DOI: 10.1007/s10557-023-07518-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/22/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a chronic vascular disease wherein the inflammation of vascular smooth muscle cells (VSMCs) plays a pivotal role in its development. Effectively mitigating AAA involves inhibiting VSMC inflammation. Agathis dammara (Lamb.) Rich, recognized for its robust anti-inflammatory and antioxidant attributes, has been employed as a traditional medicinal resource. Nonetheless, there is a dearth of information regarding the potential of Agathis dammara extract (AD) in attenuating AAA, specifically by diminishing vascular inflammation, notably VSMC inflammation. Furthermore, the active constituents of AD necessitate identification. AIM OF THE STUDY This study sought to ascertain the efficacy of AD in reducing AAA, evaluate its impact on VSMC inflammation, and elucidate whether the monomer araucarone (AO) in AD acts as an active component against AAA. MATERIALS AND METHODS The extraction of AD was conducted and subjected to analysis through High-Performance Liquid Chromatography (HPLC) and mass spectrometry. The isolation of the AO monomer followed, involving the determination of its content and purity. Subsequently, the effects of AD and AO on VSMC inflammation were assessed in vitro, encompassing an examination of inflammatory factors such as IL-6 and IL-18, as well as the activation of matrix metalloproteinase 9 (MMP9) in tumor necrosis factor-alpha (TNF-α)-stimulated VSMCs. To explore the inhibitory effects of AD/AO on AAA, C57BL/6J male mice were subjected to oral gavage (100 mg/kg) or intraperitoneal injection (50 mg/kg) of AD and AO in a porcine pancreatic elastase (PPE)-induced AAA model (14 days). This facilitated the observation of abdominal aorta dilatation, remodeling, elastic fiber disruption, and macrophage infiltration. Additionally, a three-day PPE mouse model was utilized to assess the effects of AD and AO (administered at 100 mg/kg via gavage) on acute inflammation and MMP9 expression in blood vessels. The mechanism by which AD/AO suppresses the inflammatory response was probed through the examination of NF-κB/NLRP3 pathway activation in VSMCs and aortas. RESULTS Liquid Chromatography-Mass Spectrometry (LC-MS) revealed that AO constituted 15.36% of AD's content, with a purity of 96%. Subsequent pharmacological investigations of AO were conducted in parallel with AD. Both AD and AO exhibited the ability to inhibit TNF-α-induced VSMC inflammation and MMP production in vitro. Furthermore, both substances effectively prevented PPE-induced AAA in mice, whether administered through gavage or intraperitoneal injection, evidenced by decreased vascular diameter dilation, disruption of elastin fiber layers, and infiltration of inflammatory cells. In the three-day PPE mouse model, AD and AO mitigated the heightened expression of inflammatory factors and the elevated expression of MMP9 induced by PPE. The activation of the NF-κB/NLRP3 pathway in both VSMCs and aortas was significantly suppressed by treatment with AD or AO. CONCLUSIONS Through suppressing NF-κB/NLRP3 pathway activation, AD effectively mitigates the inflammatory response in VSMCs, mitigates inflammation in aortas, prevents extracellular matrix degradation, and consequently impedes the progression of AAA. AO emerges as one of the active compounds in AD responsible for inhibiting VSMC inflammation and inhibiting AAA development.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/prevention & control
- Plant Extracts/pharmacology
- Plant Extracts/isolation & purification
- Male
- Disease Models, Animal
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/isolation & purification
- Mice, Inbred C57BL
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice
- Inflammation Mediators/metabolism
- Diterpenes/pharmacology
- Diterpenes/isolation & purification
- Cells, Cultured
- Signal Transduction
- NF-kappa B/metabolism
Collapse
Affiliation(s)
- Qingyi Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhewei Yu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Chang Di
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Yingkun Qiu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
| | - Rong Qi
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China.
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing, China.
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China.
| |
Collapse
|
20
|
Ferreira HB, Trindade F, Nogueira-Ferreira R, Leite-Moreira A, Ferreira R, Dias-Neto M, Domingues MR. Lipidomic insights on abdominal aortic aneurysm and peripheral arterial disease. J Mol Med (Berl) 2025; 103:365-380. [PMID: 40011252 PMCID: PMC12003574 DOI: 10.1007/s00109-025-02524-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 01/10/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Abdominal aortic aneurysm (AAA) and peripheral arterial disease (PAD) are two cardiovascular diseases associated with considerable morbidity, mortality and quality of life impairment. As they are multifactorial diseases, several factors contribute to their pathogenesis, including oxidative stress and lipid peroxidation, and these may have key roles in the development of these pathologies. Alterations of the lipid metabolism and lipid profile have been reported in cardiovascular diseases but to a lesser extent in AAA and PAD. Modifications in the profile of some molecular lipid species, in particular, native phospholipid and triglyceride species were mainly reported for AAA, while alterations in the fatty acid profile were noticed in the case of PAD. Oxidized phospholipids were also reported for AAA. Although AAA and PAD have a common atherosclerotic root, lipidomics demonstrates the existence of distinct lipid. Lipidomic research regarding AAA and PAD is still scarce and should be set in motion to increase the knowledge on the lipid changes that occur in these diseases, contributing not only to the discovery of new biomarkers for diagnosis and prognosis assessment but also to tailor precision medicine in the clinical field.
Collapse
Affiliation(s)
- Helena Beatriz Ferreira
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Fábio Trindade
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Rita Nogueira-Ferreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Adelino Leite-Moreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- Department of Cardiothoracic Surgery, Centro Hospitalar Universitário São João, 4200-319, Porto, Portugal
| | - Rita Ferreira
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Marina Dias-Neto
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde São João, Porto, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
21
|
Hu C, Tan H, Zhang Y, Cao G, Wu C, Lin P, Qiu S, Mo F, Wang E, Li S, Yuan T, Li Z, Fu W, Cheng D, Lai H, Chen X, Wang L. Fibroblast Activation Protein Acts as a Biomarker for Monitoring ECM Remodeling During Aortic Aneurysm via 68Ga-FAPI-04 PET Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411152. [PMID: 39950910 PMCID: PMC11984865 DOI: 10.1002/advs.202411152] [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: 09/11/2024] [Revised: 01/26/2025] [Indexed: 04/12/2025]
Abstract
Traditional imaging modalities used to monitor the diameter of aortic aneurysms (AAs) often fail to follow pathological progression. Fibroblast activation protein (FAP), a key regulator of extracellular matrix (ECM) remodeling, plays a pivotal role in aortic disease. However, its expression in the aortic wall during aneurysm progression and its potential correlation with disease severity remains unexplored. Here, utilizing histology the levels of FAP are higher in the aortic wall of patients with AA compared to healthy controls. In three distinct animal models of AA, a progressive increase in FAP expression, coincides with the advancement of ECM remodeling. Notably, the levels of 68Ga-FAPI-04 uptake in a rabbit model of abdominal AA (AAA) is positively correlated with aortic dilation (r = 0.84, p < 0.01), and the histological examination further confirmed that regions of high 68Ga-FAPI-04 uptake exhibited both increased FAP expression and more severe pathological changes. The 68Ga-FAPI-04 imaging in AA patients showed that the radiotracer specifically accumulated in the aortic walls of persistently dilated AA. These findings suggest that 68Ga-FAPI-04 positron emission tomographic (PET) imaging, by visualizing FAP localization, allows for a non-invasive approach to potentially monitor ECM remodeling during the AA progression.
Collapse
Affiliation(s)
- Chengkai Hu
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Hui Tan
- Department of Nuclear MedicineZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Yuchong Zhang
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Genmao Cao
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Chenye Wu
- Department of Cardiac SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Peng Lin
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Shouji Qiu
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Fandi Mo
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Enci Wang
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Shiyi Li
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Tong Yuan
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Zheyun Li
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Weiguo Fu
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
- Department of Vascular Surgery (Xiamen)Zhongshan hospitalFudan UniversityXiamen361015China
- Xiamen Municipal Vascular Disease Precise Diagnose & Treatment LabXiamen361015China
| | - Dengfeng Cheng
- Department of Nuclear MedicineZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Hao Lai
- Department of Cardiac SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Xiaoyuan Chen
- Departments of Diagnostic RadiologyChemical and Biomolecular Engineering, Biomedical Engineering, Pharmacy and Pharmaceutical SciencesYong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingapore119074Singapore
- Clinical Imaging Research CentreCentre for Translational MedicineYong Loo Lin School of MedicineNational University of SingaporeSingapore117599Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117597Singapore
- Theranostics Center of Excellence (TCE)Yong Loo Lin School of MedicineNational University of Singapore11 Biopolis Way, HeliosSingapore138667Singapore
- Institute of Molecular and Cell BiologyAgency for ScienceTechnology, and Research (A*STAR)61 Biopolis Drive, ProteosSingapore138673Singapore
| | - Lixin Wang
- Department of Vascular SurgeryZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
- Department of Vascular Surgery (Xiamen)Zhongshan hospitalFudan UniversityXiamen361015China
- Xiamen Municipal Vascular Disease Precise Diagnose & Treatment LabXiamen361015China
| |
Collapse
|
22
|
Zhang Z, Ling T, Ding Q, Zhu F, Cheng X, Li X, Ma T, Meng Q. GlycoRNA-rich, neutrophil membrane-coated, siMT1-loaded nanoparticles mitigate abdominal aortic aneurysm progression by inhibiting the formation of neutrophil extracellular traps. Mater Today Bio 2025; 31:101630. [PMID: 40124343 PMCID: PMC11929896 DOI: 10.1016/j.mtbio.2025.101630] [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: 12/19/2024] [Revised: 02/12/2025] [Accepted: 03/02/2025] [Indexed: 03/25/2025] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening vascular condition. Currently, there are no clinically available pharmacological interventions that can stop the progression of AAA, primarily due to the incomplete understanding of its pathogenesis and the absence of effective drug delivery systems. The present study aimed to develop a targeted therapy for AAA through a nanomedicine approach involving site-specific regulation of neutrophil extracellular trap (NET)-related pathological vascular remodeling. We found that metallothionein 1 (MT1) was upregulated in AAA lesions in both humans and mice. MT1 also facilitated the formation of NETs and subsequently induced phenotypic transformation and apoptosis in vascular smooth muscle cells. Additional in vivo studies revealed that the glycoRNA-rich membranes coated siMT1-loaded poly(lactic-co-glycolic acid) (PLGA)-polyethylene glycol (PEG) nanoparticles (GlycoRNA-NP-siMT1) effectively delivered siMT1 to AAA lesions, thereby inhibiting abdominal aortic dilation. Mechanistically, GlycoRNA-NP-siMT1 mitigated pathological remodeling of the abdominal aorta by reducing neutrophil infiltration and inhibiting the formation of NETs. Our study indicates that MT1 facilitates the progression of AAA by modulating the formation of NETs. Furthermore, GlycoRNA-NP-siMT1 show an inhibitory effect on AAA progression through a dual mechanism: they competitively inhibit neutrophil infiltration and release siMT1, which subsequently suppresses NET formation.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Tianyu Ling
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Qingwei Ding
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Feng Zhu
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiaoyuan Cheng
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiaoting Li
- Department of Geriatrics, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Teng Ma
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qingyou Meng
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
23
|
Chen D, Song T, Liu Y, Wang Y, Qin B, Zhang Q, Hu W, Zhou X, Qi R. Effective Hydrogel Vascular Patch Dual-Loaded with Cycloastragenol Nanostructured Lipid Carriers and Doxycycline for Repairing Extravascular Injury in Abdominal Aortic Aneurysm. Adv Healthc Mater 2025; 14:e2402497. [PMID: 39703126 DOI: 10.1002/adhm.202402497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/25/2024] [Indexed: 12/21/2024]
Abstract
Endovascular aneurysm repair (EVAR) plays a crucial role in the treatment of abdominal aortic aneurysm (AAA) in the clinic, but the aneurysm remains in the patient's body after surgery, continuing to pose a risk of progression. Cycloastragenol (CAG) is proven to be an effective anti-AAA drug, and its vascular protective effects can be further improved when the hydrophobic CAG is encapsulated into nano-sized formulations to enhance its bioavailability. In this context, this study developed an extravascular patch hydrogel loaded with CAG nanostructured lipid carriers and a hydrophilic drug of doxycycline hydrochloride (DOX). The extravascular patch delivered onto the mouse abdominal aortas can promote local permeation of hydrophilic/hydrophobic drugs at the vessel sites and provide effective vascular protection against AAA injury induced by elastase. This study introduces a novel and promising approach for AAA treatment, which can serve as a supplementary strategy after EVAR surgery.
Collapse
Affiliation(s)
- Du Chen
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling. NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
- School of Pharmacy / Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi, 832003, China
| | - Tiantian Song
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling. NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Yi Liu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Ying Wang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling. NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
- School of Pharmacy / Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi, 832003, China
| | - Boyang Qin
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling. NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Qingyi Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling. NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Weipeng Hu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling. NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Xiqiao Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling. NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Rong Qi
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling. NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
- School of Pharmacy / Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi, 832003, China
| |
Collapse
|
24
|
Wu Z, Wu X, Meng X, Lei J, Zeng C, Pu H, Liu Y, Xu Z, Wu X, Huang S, Qin J, Liu J, Lu X, Li B. Functional anti-inflammatory mesoporous silica nanoplatform for Synergistic and Targeted abdominal aortic aneurysm treatment. J Colloid Interface Sci 2025; 683:1040-1054. [PMID: 39721076 DOI: 10.1016/j.jcis.2024.12.140] [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: 11/26/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a chronic inflammation-driven disease characterized by aortic wall destruction and expansion, leading to high morbidity and mortality. However, previous drug treatments for its common risk factors have not achieved favorable results, and the early prevention and treatment is still the main clinical dilemma. Anti-inflammation therapy is a promising therapeutical method targeting its pathogenesis mechanism, but it has not been explored in depth. Herein, interleukin-1 receptor antagonist-loaded manganese-doped mesoporous silica nanoparticles (IL-1Ra@MMSN) were designed and synthesized to target macrophage-mediated chronic aortic inflammation for AAA treatment. IL-1Ra@MMSN showed high IL-1Ra-loading efficiency, great stability and pH-responsive drug-releasing property. IL-1Ra@MMSN specially phagocytosed by macrophages can protect against oxidative stress injury and promoted the M2 polarization via transforming growth factor-β (TGF-β) signaling in vitro. Furthermore, IL-1Ra@MMSN exhibited good lesion targeting ability, hemocompatibility and biocompatibility in angiotensin II-induced murine AAA model. In vivo experiments also confirmed the excellent treatment efficacy in reducing AAA formation and progression via protecting aortic wall integrity and promoting anti-inflammatory microenvironment. Taken together, the current study demonstrated that IL-1Ra@MMSN is a promising nanoplatform for early intervention of AAA, which provides a novel treatment strategy based on anti-inflammatory immune regulation.
Collapse
Affiliation(s)
- Zhaoyu Wu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaoyu Wu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiangtian Meng
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jiahao Lei
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Chenlin Zeng
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Hongji Pu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yijun Liu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhijue Xu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University. Shanghai 200240, China
| | - Xiaodong Wu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Sheng Huang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Jinbao Qin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jianqiang Liu
- Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, and School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Bo Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
25
|
Shi Y, Zhao Y, Sun SJ, Lan XT, Wu WB, Zhang Z, Chen YX, Yan YY, Xu YP, Li DJ, Fu H, Shen FM. Targeting GPX4 alleviates ferroptosis and retards abdominal aortic aneurysm formation. Biochem Pharmacol 2025; 234:116800. [PMID: 39952331 DOI: 10.1016/j.bcp.2025.116800] [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: 07/23/2024] [Revised: 12/16/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Abdominal aortic aneurysm (AAA) is a potentially fatal cardiovascular disease, closely related to inflammation and loss of vascular smooth muscle cells (VSMCs). Ferroptosis is an iron-dependent cell death associated with peroxidation of lipids. However, the direct role of glutathione peroxidase 4 (GPX4) itself determined ferroptosis in the course of AAA pathogenesis remains unknown. Here, we reported that ferroptosis was triggered in human AAA, elastase- and angiotensin II (Ang II)-induced mouse AAA, and Ang II-incubated VSMCs. Inhibition of ferroptosis via global genetic overexpression of GPX4, a critical anti-ferroptosis molecule, markedly prevented both vascular remodeling and inflammatory response. Mechanistically, GPX4 changed the migration and activation of macrophages/monocytes in AAA tissues in mice. Experiments in vitro demonstrated that overexpression of GPX4 prevented the JAK1/STAT3 signaling activation in VSMCs induced by IL-6, production of pro-inflammatory macrophages. Finally, the role of ferroptosis was confirmed on an Ang II-induced mice AAA model. These results emphasized the significance of ferroptosis in AAA, and provided novel insights that therapy focusing on GPX4 might be a promising strategy for treatment of AAA in the clinic.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Ferroptosis/physiology
- Animals
- Aortic Aneurysm, Abdominal/prevention & control
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
- Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors
- Phospholipid Hydroperoxide Glutathione Peroxidase/biosynthesis
- Humans
- Mice
- Mice, Inbred C57BL
- Male
- Angiotensin II/toxicity
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cells, Cultured
Collapse
Affiliation(s)
- Yu Shi
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Zhao
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Si-Jia Sun
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiu-Ting Lan
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Bin Wu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Zhen Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu-Xin Chen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu-Ying Yan
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yu-Ping Xu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Hui Fu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
26
|
Liu Y, Wang H, Yu M, Cai L, Zhao Y, Cheng Y, Deng Y, Zhao Y, Lu H, Wu X, Zhao G, Xue C, Liu H, Surakka I, Schwendeman A, Lu HS, Daugherty A, Chang L, Zhang J, Temel RE, Chen YE, Guo Y. Hypertriglyceridemia as a Key Contributor to Abdominal Aortic Aneurysm Development and Rupture: Insights from Genetic and Experimental Models. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.08.07.24311621. [PMID: 39211871 PMCID: PMC11361217 DOI: 10.1101/2024.08.07.24311621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease without effective medications. This study integrated genetic, proteomic, and metabolomic data to identify causation between increased triglyceride (TG)-rich lipoproteins and AAA risk. Three hypertriglyceridemia mouse models were employed to test the hypothesis that increased plasma TG concentrations accelerate AAA development and rupture. In the angiotensin II-infusion AAA model, most Lpl -deficient mice with severely high plasma TG concentrations died of aortic rupture. Consistently, Apoa5 -deficient mice with moderately increased TG concentrations had accelerated AAA development, while human APOC3 transgenic mice with dramatically increased TG concentrations exhibited aortic dissection and rupture. Increased TG concentrations and palmitate inhibited lysyl oxidase maturation. Locally overexpressing lysyl oxidase eliminated the impact of high TG on AAA formation in human APOC3 transgenic mice. Administration of antisense oligonucleotide targeting Angptl3 profoundly inhibited AAA progression in human APOC3 transgenic mice and Apoe -deficient mice. These results indicate that hypertriglyceridemia is a key contributor to AAA pathogenesis, highlighting the importance of triglyceride-rich lipoprotein management in treating AAA. Clinical Perspective What Is New?: This study integrates genetic, proteomic, and metabolomic data to identify causation between increased triglyceride (TG)-rich lipoproteins and AAA risk.TG concentrations influence AAA formation and severity in a dose-dependent manner, potentially by inhibiting lysyl oxidase maturation and extracellular matrix assembly. Administration of antisense oligonucleotide targeting Angptl3 profoundly inhibites AAA progression in human APOC3 transgenic mice and Apoe -deficient mice by lowering TG concentrations. What Are the Clinical Implications?: These findings underscore triglyceride-rich lipoprotein management as a promising therapeutic strategy for AAA treatment.Antisense oligonucleotide therapy targeting liver ANGPTL3 holds potential as a therapeutic approach to reduce AAA risk.
Collapse
|
27
|
Chen Q, Liu S, Zhou H, Wang J, Xiao X, Chen G, Du J, Zhong L, Song H, Huang X. SAMD4A inhibits abdominal aortic aneurysm development and VSMC phenotypic transformation through targeting KDM2B. J Adv Res 2025:S2090-1232(25)00178-X. [PMID: 40081568 DOI: 10.1016/j.jare.2025.03.018] [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: 11/29/2024] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
INTRODUCTION Abdominal aortic aneurysm (AAA) is a fatal vascular disease without effective drug treatments. Pathological vascular smooth muscle cell (VSMC) phenotypic transformation is the underlying cause of AAA. However, the underlying mechanism has not been fully elucidated. OBJECTIVE We aimed to determine whether the RNA binding protein SAMD4A suppresses VSMC phenotype transformation and inhibits AAA formation. METHODS Single-cell RNA sequencing (scRNA-seq) was conducted to reveal smooth muscle cell phenotypic heterogeneity and RNA-binding protein dysregulation during AAA formation. A pancreatic elastase (PPE)-induced mouse AAA model was generated to confirm the function of SAMD4A in vivo. RNA-seq combined with RNA immunoprecipitation (RIP) sequencing and chromatin immunoprecipitation (ChIP)-qPCR was used for mechanistic exploration. RESULTS We identified 3 smooth muscle cell subtypes, and demonstrated their transformation from contractile to inflammatory-like VSMCs during AAA formation. SAMD4A expression was increased in contractile VSMCs and significantly reduced in AAAs. The results of functional experiments revealed that VSMC-specific knockout of SAMD4A exacerbated PPE-induced AAA formation, whereas VSMC knock-in attenuated AAA formation. SAMD4A regulated VSMC contraction by binding to KDM2B. Further in vivo studies revealed that overexpression of KDM2B abolished the protective effect of SAMD4A in AAA. ChIP-qPCR demonstrated that KDM2B suppressed the transcription of VSMC contractile markers by binding to their promoters and reducing H3K4me3 and H3K36me2 levels. CONCLUSIONS SAMD4A inhibits AAA development and VSMC phenotypic transformation by targeting KDM2B. This work highlights the potential of SAMD4A as a new therapeutic option to prevent AAA formation.
Collapse
Affiliation(s)
- Qing Chen
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515 Guangdong, China; Department of Vascular Surgery, Shenzhen Third People's Hospital, the Second Affiliated Hospital, Southern University Science and Technology, China
| | - Shenrong Liu
- Department of Cardiology, the Key Laboratory of Advanced Interdisciplinary Studies Center, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120 Guangdong, China
| | - Haobin Zhou
- Department of Cardiology, the Key Laboratory of Advanced Interdisciplinary Studies Center, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120 Guangdong, China
| | - Junfen Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyong Xiao
- Department of Emergency Medicine, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, China
| | - Guojun Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou 510515, China
| | - Juan Du
- Department of Geriatrics, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Lintao Zhong
- Department of Cardiology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, China.
| | - Haoyu Song
- Department of Geriatrics, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China.
| | - Xianying Huang
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515 Guangdong, China.
| |
Collapse
|
28
|
Qi Y, Jiang H, Lun Y, Gang Q, Shen S, Zhang H, Liu M, Wang Y, Zhang J. Protein Drug Targets for Abdominal Aortic Aneurysm and Proteomic Associations Between Modifiable Risk Factors and Abdominal Aortic Aneurysm. J Am Heart Assoc 2025; 14:e037802. [PMID: 40008516 DOI: 10.1161/jaha.124.037802] [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: 08/05/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a severe aortic disease for which no pharmacological interventions have yet been developed. This investigation focused on identifying protein-based therapeutic targets and assessing how proteins mediate the interplay between modifiable risk factors and AAA development. METHODS Causal inferences between plasma proteins and AAA were drawn using 2-sample Mendelian randomization, followed by comprehensive sensitivity testing, colocalization, and replication efforts. Further analyses included database interrogation, single-cell RNA data analysis, enrichment analysis, protein-protein interaction networks, and immunohistochemistry to map the tissue-specific expression of these proteins, their expression within AAA tissues, and their biological roles. Mediation Mendelian randomization was employed to evaluate the mediating effects of AAA-related proteins on the associations between AAA and 3 risk factors: hypertension, smoking, and obesity. RESULTS A total of 43 proteins were identified as having causal links to AAA. Colocalization analysis pinpointed 13 proteins with strong evidence of colocalization with AAA. Of these, the causal involvement of 10 proteins was substantiated by external validation data. Consistent evidence for PCSK9 (proprotein convertase subtilisin/kexin type 9), IL6R (interleukin-6R), ECM1 (extracellular matrix protein 1), and ANGPTL4 (angiopoietin-related protein 4) was further validated through tissue immunohistochemistry and blood data. Moreover, Mendelian randomization analysis identified 10 proteins as mediators of the influence of hypertension, smoking, and obesity on AAA development. CONCLUSIONS This analysis identifies 4 proteins (PCSK9, IL6R, ECM1, and ANGPTL4) as high-priority therapeutic targets for AAA and emphasizes the intermediary role of plasma proteins in linking hypertension, smoking, obesity, and AAA. Further investigations are needed to clarify the specific roles of these proteins in AAA pathology.
Collapse
Affiliation(s)
- Yao Qi
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Han Jiang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Yu Lun
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Qingwei Gang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Shikai Shen
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Han Zhang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Mingyu Liu
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Yixian Wang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Jian Zhang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| |
Collapse
|
29
|
Kucher AN, Koroleva IA, Nazarenko MS. Exploring Disparities in Atherosclerosis Comorbidity with Aortic Aneurysm. Biomedicines 2025; 13:593. [PMID: 40149570 PMCID: PMC11940622 DOI: 10.3390/biomedicines13030593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 03/29/2025] Open
Abstract
Aortic aneurysm (AA) and atherosclerosis (AS) of various vascular beds are asymptomatic for a long time and are relatively common pathological conditions that lead to life-threatening and disabling complications. In this review, we discuss the current understanding of the high variation in direct and inverse comorbidity of AA and AS as presented in scientific publications. Estimates of AA and AS comorbidity depend on several factors, such as the location of AA (ascending or descending thoracic aorta or abdominal aorta), familial or sporadic cases of AA, syndromic forms of AA, and/or aortic valve pathology (bicuspid aortic valve [BAV]). To identify the causes of the comorbidity of AA and AS, it is important to consider and characterise many factors in detail. These factors include clinical characteristics of the patients included in a study (age, sex) and risk factors (mainly the presence of monogenic forms and BAV, hypertension, hypercholesterolaemia, diabetes mellitus, and cigarette smoking). Additionally, it is essential to consider characteristics of the disease course and the nature of multimorbidity and to take into account pathologies not only of the cardiovascular system but also of other organ systems, with special attention to metabolic and endocrine disorders.
Collapse
Affiliation(s)
| | | | - Maria S. Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| |
Collapse
|
30
|
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
|
31
|
Puertas-Umbert L, Alonso J, Blanco-Casoliva L, Almendra-Pegueros R, Camacho M, Rodríguez-Sinovas A, Galán M, Roglans N, Laguna JC, Martínez-González J, Rodríguez C. Inhibition of ATP-citrate lyase by bempedoic acid protects against abdominal aortic aneurysm formation in mice. Biomed Pharmacother 2025; 184:117876. [PMID: 39889383 DOI: 10.1016/j.biopha.2025.117876] [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: 11/20/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
Abdominal aortic aneurysm (AAA) is a prevalent degenerative disease characterized by an exacerbated inflammation and destructive vascular remodeling. Unfortunately, effective pharmacological tools for the treatment of this disease remain a challenge. ATP-citrate lyase (ACLY), the primary enzyme responsible for acetyl-CoA biosynthesis, is a key regulator of inflammatory signaling in macrophages and lymphocytes. Here, we found increased levels of the active (phosphorylated) form of ACLY (p-ACLY) in the inflammatory infiltrate of AAA from patients and in aneurysmal lesions from angiotensin II (Ang II)-infused apolipoprotein E-deficient mice (ApoE-/-). Furthermore, plasma ACLY levels positively correlates with IL6 and IFNγ levels in patients with AAA, while inflammatory stimuli strongly upregulated ACLY expression in macrophages and Jurkat cells. The administration of the ACLY inhibitor bempedoic acid (BemA) protected against Ang II-induced AAA formation in ApoE-/- mice, limiting the progression of aortic dilatation and reducing mortality due to aortic rupture. BMS-303141, another ACLY inhibitor, also ameliorated AAA formation, although to a lesser extent. BemA attenuated vascular remodeling and the disorganization and rupture of elastic fibers induced by Ang II, as well as vascular inflammation, decreasing the recruitment of macrophages (CD68 +) and neutrophils (Ly-6G+) into the aortic wall. Moreover, BemA shifted splenic monocytes toward a functionally anti-inflammatory phenotype, and increased the percentage of CD4+CD69+ cells. Taken together, these results support the contribution of ACLY to AAA and point to BemA as a promising tool to be considered for future clinical trials addressing the management of this disease which is quite often associated with disorders of lipoprotein metabolism.
Collapse
Affiliation(s)
- Lídia Puertas-Umbert
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Laia Blanco-Casoliva
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain
| | | | - Mercedes Camacho
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Antonio Rodríguez-Sinovas
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain; Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Barcelona 08035, Spain
| | - María Galán
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain; Facultad de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Madrid 28922, Spain
| | - Nuria Roglans
- Dept. Farmacologia, Toxicologia i Química Terapèutica. Facultat de Farmàcia i Ciències de l'Alimentació, Institut de Biomedicina, Universitat de Barcelona, Barcelona 08028, Spain; CIBER de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Juan Carlos Laguna
- Dept. Farmacologia, Toxicologia i Química Terapèutica. Facultat de Farmàcia i Ciències de l'Alimentació, Institut de Biomedicina, Universitat de Barcelona, Barcelona 08028, Spain; CIBER de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Cristina Rodríguez
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain.
| |
Collapse
|
32
|
van Merrienboer TAR, Rombouts KB, Bogunovic N, Mieremet A, Meekel JP, Balm R, de Waard V, Yeung KK. Metformin Improves the Function of Abdominal Aortic Aneurysm Patient-Derived Aortic Smooth Muscle Cells. Eur J Vasc Endovasc Surg 2025; 69:485-495. [PMID: 39321955 DOI: 10.1016/j.ejvs.2024.09.022] [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: 11/10/2023] [Revised: 08/06/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) is a cardiovascular risk factor. Paradoxically, a decreased risk of abdominal aortic aneurysm (AAA) presence and growth rate is described among patients with T2DM, associated with metformin use. This study aimed to investigate the effect of metformin on AAA patient-derived aortic smooth muscle cell (SMC) function. METHODS Aortic biopsies were obtained from patients with AAA (n = 21) and controls (n = 17) during surgery. The SMCs of non-pathological aortic controls, non-diabetic patients with AAA, and diabetic patients with AAA were cultured from explants and treated with or without metformin. The SMC contractility was measured upon ionomycin stimulation, as well as metabolic activity, proliferation, and migration. mRNA and protein expression of markers for contraction, metabolic activity, proliferation, and inflammation were measured. RESULTS mRNA expression of KLF4 and GYS1, genes involved in metabolic activity, differed between SMCs from non-diabetic and diabetic patients with AAA before metformin stimulation (p < .041). However, the effect of metformin on the various SMC functions was similar between non-diabetic and diabetic patients with AAA. Upon stimulation, metformin increased the contractility of AAA patient SMCs (p = .001). mRNA expression of smoothelin, a marker for the contractile phenotype, increased in SMCs of patients with AAA after treatment with metformin (p = .006). An increase in metabolic activity (p < .001) and a decrease in proliferation (p < .001) and migration were found in the SMCs of controls and patients with AAA with metformin. Increased mRNA expression of PPARγ, a nuclear receptor involved in mitochondrial biogenesis (p < .009), and a decrease in gene expression of Ki-67, a marker for proliferation (p < .005), were observed. Gene expression of inflammation markers MCP-1 and IL-6, and protein expression of NF-κB p65 decreased after treatment with metformin in patients with AAA. CONCLUSION This study found that metformin increases contractility and metabolic activity, and reduces proliferation, migration, and inflammation in aortic SMCs in vitro.
Collapse
MESH Headings
- Humans
- Metformin/pharmacology
- Metformin/therapeutic use
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/genetics
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Kruppel-Like Factor 4
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cell Proliferation/drug effects
- Aged
- Female
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/complications
- Hypoglycemic Agents/pharmacology
- Cell Movement/drug effects
- Cells, Cultured
- Middle Aged
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Case-Control Studies
- Muscle Contraction/drug effects
Collapse
Affiliation(s)
- Tara A R van Merrienboer
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Physiology, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, the Netherlands.
| | - Karlijn B Rombouts
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Physiology, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, the Netherlands
| | - Natalija Bogunovic
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands
| | - Arnout Mieremet
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, the Netherlands
| | - Jorn P Meekel
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands
| | - Ron Balm
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands
| | - Vivian de Waard
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, the Netherlands
| | - Kak K Yeung
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Physiology, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, the Netherlands
| |
Collapse
|
33
|
Ye W, Yang P, Jin M, Zou J, Zheng Z, Li Y, Zhang D, Ye W, Huang Z, Wang J, Liu Z. Dihydromyricetin mitigates abdominal aortic aneurysm via transcriptional and post-transcriptional regulation of heme oxygenase-1 in vascular smooth muscle cells. Acta Pharm Sin B 2025; 15:1514-1534. [PMID: 40370559 PMCID: PMC12069254 DOI: 10.1016/j.apsb.2025.02.003] [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: 07/10/2024] [Revised: 09/15/2024] [Accepted: 11/26/2024] [Indexed: 05/16/2025] Open
Abstract
Abdominal aortic aneurysm (AAA) is a deadly condition of the aorta, carrying a significant risk of death upon rupture. Currently, there is a dearth of efficacious pharmaceutical interventions to impede the advancement of AAA and avert it from rupturing. Here, we investigated dihydromyricetin (DHM), one of the predominant bioactive flavonoids in Ampelopsis grossedentata (A. grossedentata), as a potential agent for inhibiting AAA. DHM effectively blocked the formation of AAA in angiotensin II-infused apolipoprotein E-deficient (ApoE-/-) mice. A combination of network pharmacology and whole transcriptome sequencing analysis revealed that DHM's anti-AAA action is linked to heme oxygenase (HO)-1 (Hmox-1 for the rodent gene) and hypoxia-inducible factor (HIF)-1α in vascular smooth muscle cells (VSMCs). Remarkably, DHM caused a robust rise (∼10-fold) of HO-1 protein expression in VSMCs, thereby suppressing VSMC inflammation and oxidative stress and preserving the VSMC contractile phenotype. Intriguingly, the therapeutic effect of DHM on AAA was largely abrogated by VSMC-specific Hmox1 knockdown in mice. Mechanistically, on one hand, DHM increased the transcription of Hmox-1 by triggering the nuclear translocation and activation of HIF-1α, but not nuclear factor erythroid 2-related factor 2 (NRF2). On the other hand, molecular docking, combined with cellular thermal shift assay (CETSA), isothermal titration calorimetry (ITC), drug affinity responsive target stability (DARTS), co-immunoprecipitation (Co-IP), and site mutant experiments revealed that DHM bonded to HO-1 at Lys243 and prevented its degradation, thereby resulting in considerable HO-1 buildup. In summary, our findings suggest that naturally derived DHM has the capacity to markedly enhance HO-1 expression in VSMCs, which may hold promise as a therapeutic strategy for AAA.
Collapse
Affiliation(s)
- Weile Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Mei Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Jiami Zou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Zhihua Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Yuanyuan Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Wencai Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Zunnan Huang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jiaojiao Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| |
Collapse
|
34
|
Soudant J, González-Blázquez R, Merino A, Ballesteros-Martínez C, Rodrigues-Diez R, Moreno-Carriles R, Nistal JF, Guerra S, Redondo JM, Salaices M, Briones AM, García-Redondo AB. Interferon stimulated gene 15 (ISG15) modulates phenotype of vascular smooth muscle cells and pathological vascular remodeling. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2025:500769. [PMID: 40023687 DOI: 10.1016/j.arteri.2025.500769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 03/04/2025]
Abstract
INTRODUCTION Inflammation is a major determinant of abdominal aortic aneurysms (AAA). Interferon stimulated gene 15 (ISG15) has a role in vascular remodelling in AAA. This study investigates the mechanisms whereby ISG15 might affect vascular remodeling and function. METHODS We used vascular smooth muscle cells (VSMC) from wild type (ISG15+/+) o ISG15 knockout (ISG15-/-) mice, aorta from ISG15+/+ and ISG15-/- mice infused with angiotensin II (1.44mg/kg/day, sc, 14 days), and human AAA. We also performed a model of recombinant ISG15 infusion (rISG15, sc, 100 and 500ng/day, 14 days) in mice. RESULTS In VSMC, ISG15 deficiency increased the expression of contractile (Acta2, Tagln) and synthetic (Fn1, Col1a2, Col3, Col4) markers and decreased the expression of the calcification marker Spp1. Ang II infusion changed the expression of phenotype markers differently in aorta from ISG15+/+ or ISG15-/- mice. ISG15 expression showed a negative correlation with expression of contractile markers (ACTA2, CNN1), and with COL3a1, in human samples from patients with AAA or with stenotic aorto-iliac pathology. rISG15 infusion induced hypotrophic vascular remodelling in mesenteric arteries without affecting vascular mechanics. Aorta of ISG15-/- mice contracted more to thromboxane A2 analogue U46619, compared to ISG15-/-mice. Both aorta and mesenteric arteries from rISG15-treated mice showed less contractility than control mice. CONCLUSIONS ISG15 participates in pathological vascular remodeling probably by modulating VSMC phenotype. These changes could also impact in the vascular function.
Collapse
Affiliation(s)
- Julius Soudant
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España
| | - Raquel González-Blázquez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España
| | - Abraham Merino
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España
| | - Constanza Ballesteros-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España; CIBER Cardiovascular, España
| | - Raquel Rodrigues-Diez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España; CIBER Cardiovascular, España
| | - Rosa Moreno-Carriles
- Servicio de Angiología y Cirugía Vascular, Hospital Universitario La Princesa, Madrid, España
| | - J Francisco Nistal
- CIBER Cardiovascular, España; Cirugía Cardiovascular, Hospital Universitario Marqués de Valdecilla, IDIVAL, Departamento de Ciencias Médica y Quirúrgicas, Facultad de Medicina, Universidad de Cantabria, Santander, España
| | - Susana Guerra
- Departamento de Medicina Preventiva y Microbiología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España
| | - Juan Miguel Redondo
- CIBER Cardiovascular, España; Grupo de Regulación Génica en Remodelado Cardiovascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares, Madrid, España; Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas,Universidad Autónoma de Madrid, Madrid, España
| | - Mercedes Salaices
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España; CIBER Cardiovascular, España
| | - Ana M Briones
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España; CIBER Cardiovascular, España.
| | - Ana B García-Redondo
- CIBER Cardiovascular, España; Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España.
| |
Collapse
|
35
|
Tian Z, Li W, Wang J, Li S. WTAP-mediated m6A modification on BASP1 mRNA contributes to ferroptosis in AAA. Gen Thorac Cardiovasc Surg 2025:10.1007/s11748-025-02130-5. [PMID: 39969669 DOI: 10.1007/s11748-025-02130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a common aneurysm that is often associated with atherosclerosis and can lead to artery rupture and death. Brain abundant membrane attached signal protein 1 (BASP1) is related to a variety of pathophysiological processes, but its role in AAA has not been reported. METHODS Real-time quantitative polymerase chain reaction (qRT-PCR) and western blot were used to detect the expressions of BASP1 and Wilms' tumor 1-associated protein (WTAP). Angiotensin-II (Ang-II) was employed for inducing AAA models in vitro to explore the effects and mechanism of BASP1 in AAA. Cell viability, apoptosis, oxidative stress level, and Fe2+ level were measured by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl tetrazolium bromide (MTT), flow cytometry, and various kits, respectively. In terms of mechanism, the methylated RNA immunoprecipitation (MeRIP)-qPCR, the dual luciferase reporter assay, and the cytochrome experiments were utilized to evaluate the relationship between BASP1 and WTAP. RESULTS A highly expressed level of BASP1 was observed in aortic tissues of AAA patients and Ang-II could induce AAA models by treating vascular smooth muscle cells (VSMCs). In cellular function, BASP1 knockdown impaired AAA and ferroptosis resulted from Ang-II. Mechanically, WTAP mediated the N6-methyladenosine (m6A) modification and mRNA stability of BASP1. Meanwhile, WTAP was highly expressed in AAA tissues of patients and the effects of WTAP silence in AAA and ferroptosis were diminished by up-regulated BASP1. CONCLUSION WTAP promotes cell viability and inhibits apoptosis and ferroptosis resulted from Ang-II in VSMCs by mediating the m6A level of BASP1.
Collapse
Affiliation(s)
- Zexiang Tian
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, No.1, Tongdao North Street, Huimin District, Hohhot, 010050, China
| | - Wei Li
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, No.1, Tongdao North Street, Huimin District, Hohhot, 010050, China
| | - Jian Wang
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, No.1, Tongdao North Street, Huimin District, Hohhot, 010050, China
| | - Shuzhen Li
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, No.1, Tongdao North Street, Huimin District, Hohhot, 010050, China.
| |
Collapse
|
36
|
Zhang Q, Ding H, Dai Z, Yang R, Zhou S, Tai S. U-shaped association between plasma cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) levels and myocardial infarction. BMC Cardiovasc Disord 2025; 25:116. [PMID: 39972291 PMCID: PMC11837390 DOI: 10.1186/s12872-025-04543-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/31/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND The cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) signaling pathway is closely associated with myocardial infarction (MI). Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) is a key component of this pathway; however, there is currently a lack of clinical evidence linking plasma cGAMP levels to MI. METHODS This study utilized clinical data from 270 patients diagnosed with coronary heart disease (CHD) at the Second Xiangya Hospital of Central South University. The outcomes included ST-segment elevation and non-ST-segment elevation MI. Univariate and multivariate logistic regression models were used to explore the relationships between plasma cGAMP levels and MI, while restricted cubic spline (RCS) using logistic regression to explore the dose-response relationship. RESULTS Among the 270 patients, the mean plasma cGAMP level was 1352.58 ± 106.02 ng/L and 89 (32.96%) patients were diagnosed with MI. The RCS curves indicated a U-shape association between the cGAMP levels and MI; the risk of MI was negatively correlated with the cGAMP until it hit bottoms at 1352 ng/L. When the cGAMP level exceeded 1352 ng/L, the risk of MI increased significantly (adjusted OR, 1.02; 95% CI: 1.01-1.03). When considering cGAMP as a categorical variable, patients in Tertile 1 and Tertile 3 had a 167% (adjusted OR: 2.67, 95% CI: 1.23-5.78) and 155% (adjusted OR: 2.55, 95% CI: 1.17-5.55) higher risk of MI compared to those in Tertile 2, respectively. These results were consistent across subgroup analyses, notably, a significant interaction by age category was observed in patients with cGAMP ≥ 1352 ng/L, where the positive association was pronounced in the elderly. CONCLUSIONS A U-shaped association exists between cGAMP and MI in the CHD population, with a cutoff point at the cGAMP of 1352 ng/L. Both excessively high and low cGAMP levels are associated with an increased risk of MI, particularly among the elderly with cGAMP ≥ 1352 ng/L. This is the first clinical evidence of the cGAS-cGAMP-STING pathway in metabolic cardiovascular diseases. CLINICALTRIALS GOV IDENTIFIER NCT03363035 (Registration date: 2018-01-15).
Collapse
Affiliation(s)
- Quan Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Huiqing Ding
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Zhongling Dai
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Rukai Yang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shi Tai
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
37
|
Xu C, Wang G, Jin G, Fei X, Liu C, Tang L, Fu L, Yu J. Genetic association between inflammatory factors and abdominal aortic aneurysm: Insights from a genome-wide association study. Int J Cardiol 2025; 421:132905. [PMID: 39662749 DOI: 10.1016/j.ijcard.2024.132905] [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: 03/28/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a fatal vascular disorder. The current primary treatment for AAA remains restricted to surgical intervention during advanced stages of the disease, with no efficacious pharmaceutical options available for early-stage AAA patients. Inflammation is known to play a substantial role in the development of AAA, with various inflammatory factors implicated in its pathogenesis. However, conflicting findings have been reported in studies investigating the roles of these inflammatory factors in AAA, making it challenging to establish definitive causal relationships between inflammatory factors and AAA. METHODS The research conducted a bidirectional Mendelian randomization (MR) study using genetic variants. Inflammatory factors were obtained from a genome-wide association study (GWAS), while AAA were sourced from the FinnGen consortium. The primary method employed was inverse-variance weighted (IVW), with MR-Egger, weighted median, and MR-PRESSO approaches used as supplementary analyses. RESULTS According to the IVW method, hepatocyte growth factor (HGF), matrix metalloproteinase-7 (MMP-7), MMP-12, and NF-kappa-B essential modulator (NEMO/ IKKγ) were associated with a potential increased risk of AAA, while platelet-derived growth factor BB (PDGFbb), interleukin-4 (IL-4), IL-12p70, IL-10, IL-6Rα, and myeloperoxidase (MPO) were associated with a potential decreased risk of AAA. In the reverse MR analysis, no causal relationship was observed between AAA and any of the inflammatory factors. CONCLUSIONS This study provides evidence supporting a causal relationship between inflammatory factors and AAA. It suggests that targeting and modulating these specific inflammatory factors may serve as a potential approach for the prevention and noninvasive treatment of AAA.
Collapse
Affiliation(s)
- Chao Xu
- Department of Vascular and Hernia Surgery, Shaoxing People's Hospital, Shaoxing City, Zhejiang Province, PR China
| | - Guohua Wang
- Department of Vascular and Hernia Surgery, Shaoxing People's Hospital, Shaoxing City, Zhejiang Province, PR China
| | - Gan Jin
- Department of Vascular and Hernia Surgery, Shaoxing People's Hospital, Shaoxing City, Zhejiang Province, PR China
| | - Xiaozhou Fei
- Department of Vascular and Hernia Surgery, Shaoxing People's Hospital, Shaoxing City, Zhejiang Province, PR China
| | - Chunjiang Liu
- Department of Vascular and Hernia Surgery, Shaoxing People's Hospital, Shaoxing City, Zhejiang Province, PR China
| | - Liming Tang
- Department of Vascular and Hernia Surgery, Shaoxing People's Hospital, Shaoxing City, Zhejiang Province, PR China
| | - Leihua Fu
- Department of Hematology, Shaoxing People's Hospital, Shaoxing City, Zhejiang Province, PR China
| | - Jieni Yu
- Department of Hematology, Shaoxing People's Hospital, Shaoxing City, Zhejiang Province, PR China.
| |
Collapse
|
38
|
Rijken L, Zwetsloot S, Smorenburg S, Wolterink J, Išgum I, Marquering H, van Duivenvoorde J, Ploem C, Jessen R, Catarinella F, Lee R, Bera K, Buisan J, Zhang P, Dias-Neto M, Raffort J, Lareyre F, Muller C, Koncar I, Tomic I, Živković M, Djuric T, Stankovic A, Venermo M, Tulamo R, Behrendt CA, Smit N, Schijven M, van den Born BJ, Delewi R, Jongkind V, Ayyalasomayajula V, Yeung KK. Developing Trustworthy Artificial Intelligence Models to Predict Vascular Disease Progression: the VASCUL-AID-RETRO Study Protocol. J Endovasc Ther 2025:15266028251313963. [PMID: 39921236 DOI: 10.1177/15266028251313963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2025]
Abstract
INTRODUCTION Abdominal aortic aneurysms (AAAs) and peripheral artery disease (PAD) are two vascular diseases with a significant risk of major adverse cardiovascular events and mortality. A challenge in current disease management is the unpredictable disease progression in individual patients. The VASCUL-AID-RETRO study aims to develop trustworthy multimodal predictive artificial intelligence (AI) models for multiple tasks including risk stratification of disease progression and cardiovascular events in patients with AAA and PAD. METHODS The VASCUL-AID-RETRO study will collect data from 5000 AAA and 6000 PAD patients across multiple European centers of the VASCUL-AID consortium using electronic health records from 2015 to 2024. This retrospectively-collected data will be enriched with additional data from existing biobanks and registries. Multimodal data, including clinical records, radiological imaging, proteomics, and genomics, will be collected to develop AI models predicting disease progression and cardiovascular risks. This will be done while integrating the international ethics guidelines and legal standards for trustworthy AI, to ensure a socially-responsible data integration and analysis. PROPOSED ANALYSES A consensus-based variable list of clinical parameters and core outcome set for both diseases will be developed through meetings with key opinion leaders. Blood, plasma, and tissue samples from existing biobanks will be analyzed for proteomic and genomic variations. AI models will be trained on segmented AAA and PAD artery geometries for estimation of hemodynamic parameters to quantify disease progression. Initially, risk prediction models will be developed for each modality separately, and subsequently, all data will be combined to be used as input to multimodal prediction models. During all processes, data security, data quality, and ethical guidelines and legal standards will be carefully considered. As a next step, the developed models will be further adjusted with prospective data and internally validated in a prospective cohort (VASCUL-AID-PRO study). CONCLUSION The VASCUL-AID-RETRO study will utilize advanced AI techniques and integrate clinical, imaging, and multi-omics data to predict AAA and PAD progression and cardiovascular events. CLINICAL TRIAL REGISTRATION The VASCUL-AID-RETRO study is registered at www.clinicaltrials.gov under the identification number NCT06206369. CLINICAL IMPACT The VASCUL-AID-RETRO study aims to improve clinical practice of vascular surgery by developing artificial intelligence-driven multimodal predictive models for patients with abdominal aortic aneurysms or peripheral artery disease, enhancing personalized medicine. By integrating comprehensive data sets including clinical, imaging, and multi-omics data, these models have the potential to provide accurate risk stratification for disease progression and cardiovascular events. An innovation lies in the extensive European data set in combination with multimodal analyses approaches, which enables the development of advanced models to facilitate better understanding of disease mechanisms and progression. For clinicians, this means that more precise, individualized treatment plans can be established, ultimately aiming to improve patient outcomes.
Collapse
Affiliation(s)
- Lotte Rijken
- Department of Surgery, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
- Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Digital Health Amsterdam Public Health, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Sabrina Zwetsloot
- Department of Surgery, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
- Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Stefan Smorenburg
- Department of Surgery, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
- Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jelmer Wolterink
- Department of Applied Mathematics, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Ivana Išgum
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands
- Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Henk Marquering
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands
| | - Jan van Duivenvoorde
- Department of Surgery, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
- Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Corrette Ploem
- Department of Ethics, Law and Humanities, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands
| | - Roosmarie Jessen
- Department of Ethics, Law and Humanities, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands
| | | | - Regent Lee
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Katarzyna Bera
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Jenny Buisan
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Ping Zhang
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Marina Dias-Neto
- Department of Angiology and Vascular Surgery, Centro Hospitalar Universitário de São João, Porto, Portugal
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Juliette Raffort
- Clinical Chemistry Laboratory, University Hospital of Nice, Nice, France
- Institute 3IA Côte d'Azur, Université Côte d'Azur, Nice, France
- CNRS, UMR7370, LP2M, Université Côte d'Azur, Nice, France
| | - Fabien Lareyre
- CNRS, UMR7370, LP2M, Université Côte d'Azur, Nice, France
- Department of Vascular Surgery, Hospital of Antibes Juan-les-Pins, Antibes, France
| | | | - Igor Koncar
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Ivan Tomic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Maja Živković
- Laboratory for Radiobiology and Molecular Genetics, VINCA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Djuric
- Laboratory for Radiobiology and Molecular Genetics, VINCA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, VINCA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Maarit Venermo
- Department of Vascular Surgery, Helsinki University Hospital, Helsinki, Finland
- Department of Vascular Surgery, University of Helsinki, Helsinki, Finland
| | - Riikka Tulamo
- Department of Vascular Surgery, Helsinki University Hospital, Helsinki, Finland
- Department of Vascular Surgery, University of Helsinki, Helsinki, Finland
| | - Christian-Alexander Behrendt
- Department of Vascular and Endovascular Surgery, Asklepios Clinic Wandsbek, Asklepios Medical School, Hamburg, Germany
| | - Noeska Smit
- Department of Informatics, University of Bergen, Bergen, Norway
- Department of Radiology, Mohn Medical Imaging and Visualization Centre, Haukeland University Hospital, Bergen, Norway
| | - Marlies Schijven
- Digital Health Amsterdam Public Health, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Bert-Jan van den Born
- Department of Public and Occupational Health, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands
- Department of Vascular Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands
| | - Ronak Delewi
- Department of Cardiology, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Vincent Jongkind
- Department of Surgery, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
- Microcirculation, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Venkat Ayyalasomayajula
- Department of Surgery, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
- Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
- Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Wang X, Kuang J, Li XT, Hu X, Liu YH, Hu CP, Wang M, Wang Q, Zhang Z. Dimethyl fumarate is repurposed to ameliorate aortic aneurysm and dissection in mice. Eur J Pharmacol 2025; 988:177215. [PMID: 39706468 DOI: 10.1016/j.ejphar.2024.177215] [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: 07/25/2024] [Revised: 11/21/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Aortic aneurysm and dissection pose fatal threats but no effective drug therapies are available. Previous work has been directed to reduce risk factors or target key pathological events, but none of the translational efforts succeeds. Here, we attempt to repurpose dimethyl fumarate (DMF), an FDA-approved immunomodulatory drug for multiple sclerosis, for the treatment of aortic aneurysm and dissection. In three preclinical mouse models of abdominal aortic aneurysm (porcine pancreatic elastase perfusion or CaCl2 incubation) and thoracic aortic aneurysm and dissection (β-Aminopropionitrile feeding), DMF invariably protected mice from aneurysm growth, aortic dissection, rupture and death. Histological H&E and EVG staining demonstrated aortic architecture-preserving effects of DMF. Through transcriptome profiling and the connectivity map (CMap), we showed that DMF restored SRC-FAK signaling in aortic smooth muscle cells and increased collagen I turnover in the tunica media. Our work suggests the potential of DMF being repurposed for aortic aneurysm and dissection, and highlights the importance of SRC-FAK signaling in aortic homeostasis.
Collapse
MESH Headings
- Animals
- Dimethyl Fumarate/pharmacology
- Dimethyl Fumarate/therapeutic use
- Aortic Dissection/drug therapy
- Aortic Dissection/pathology
- Aortic Dissection/metabolism
- Aortic Dissection/chemically induced
- Mice
- Drug Repositioning
- Male
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/drug therapy
- Aortic Aneurysm, Thoracic/chemically induced
- Disease Models, Animal
- Mice, Inbred C57BL
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/chemically induced
- Signal Transduction/drug effects
- src-Family Kinases/metabolism
- Aortic Aneurysm/drug therapy
- Aortic Aneurysm/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Focal Adhesion Protein-Tyrosine Kinases/metabolism
Collapse
Affiliation(s)
- Xuan Wang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Jin Kuang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Xiao-Tian Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Xi Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Yu-Hang Liu
- Department of the Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Chang-Ping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, Hunan, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, 410078, Hunan, China
| | - Mi Wang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qing Wang
- Department of the Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China.
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, Hunan, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
40
|
Zhu Q, Luo D, Li Y, Yu L, Zhang Z, Ouyang F, Li L, Lu M, Hu C, Dong Y, Ma C, Liang Y, Zhao TJ, Chen FJ, Li P, Yang TS. CIDEC/FSP27 exacerbates obesity-related abdominal aortic aneurysm by promoting perivascular adipose tissue inflammation. LIFE METABOLISM 2025; 4:loae035. [PMID: 39872985 PMCID: PMC11770823 DOI: 10.1093/lifemeta/loae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 08/23/2024] [Accepted: 09/17/2024] [Indexed: 01/30/2025]
Abstract
Abdominal aortic aneurysm (AAA) is strongly correlated with obesity, partially due to the abnormal expansion of abdominal perivascular adipose tissue (PVAT). Cell death-inducing DNA fragmentation factor-like effector C (CIDEC), also known as fat-specific protein 27 (FSP27) in rodents, is specifically expressed in adipose tissue where it mediates lipid droplet fusion and adipose tissue expansion. Whether and how CIDEC/FSP27 plays a role in AAA pathology remains elusive. Here, we show that FSP27 exacerbates obesity and angiotensin Ⅱ (Ang Ⅱ)-induced AAA progression. FSP27 deficiency in mice inhibited high-fat diet-induced PVAT expansion and inflammation. Both global and adipose tissue-specific FSP27 ablation significantly decreased obesity-related AAA incidence. Deficiency of FSP27 in adipocytes abrogated matrix metalloproteinase-12 (MMP12) expression in aortic tissues. Infiltrated macrophages, which partially colocalize with MMP12, were significantly decreased in the FSP27-deficient aorta. Mechanistically, knockdown of Fsp27 in 3T3-L1 adipocytes inhibited C-C motif chemokine ligand 2 (CCL2) expression and secretion through a c-Jun N-terminal kinase (JNK)-dependent pathway, thereby leading to reduced induction of macrophage migration, while Cidec overexpression rescued this effect. Overall, our study demonstrates that CIDEC/FSP27 in adipose tissue contributes to obesity-related AAA formation, at least in part, by enhancing PVAT inflammation and macrophage infiltration, thus shedding light on its significance as a key regulator in the context of obesity-related AAA.
Collapse
Affiliation(s)
- Qing Zhu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Da Luo
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Shanghai Qi Zhi Institute, Shanghai 200030, China
| | - Yining Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Liyang Yu
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100086, China
| | - Zixuan Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Feng Ouyang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Liangkui Li
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100086, China
| | - Manxi Lu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Changyong Hu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Yinuo Dong
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Chengxin Ma
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Yan Liang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Tong-Jin Zhao
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Feng-Jung Chen
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Shanghai Qi Zhi Institute, Shanghai 200030, China
| | - Peng Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Shanghai Qi Zhi Institute, Shanghai 200030, China
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100086, China
- School of Life Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Tian-Shu Yang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Shanghai Qi Zhi Institute, Shanghai 200030, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
41
|
Guo M, Ji S, Wang H, Zhang J, Zhu J, Yang G, Chen L. Myeloid Cell mPGES-1 Deletion Attenuates Calcium Phosphate-induced Abdominal Aortic Aneurysm in Male Mice. Inflammation 2025; 48:288-298. [PMID: 38865056 DOI: 10.1007/s10753-024-02055-w] [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: 10/18/2023] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 06/13/2024]
Abstract
Microsomal PGE2 synthase (mPGES)-1 is the key enzyme responsible for synthesizing inflammatory prostaglandin E2 (PGE2). Our previous studies have shown that deletion mPGES-1 in myeloid cells hinders atherogenesis, suppresses vascular proliferative response to injury and enhances survival after myocardial infarction. Here we aimed to further explore the influence of myeloid cell mPGES-1 deletion in abdominal aortic aneurysm (AAA) formation. The AAA was triggered by applying 0.5 M calcium phosphate (CaPO4) to the infrarenal aorta of both myeloid mPGES-1 knockout (Mac-mPGES-1-KO) and their littermate control Mac-mPGES-1-WT mice. AAA induction was assessed by calculating the expansion of the infrarenal aortic diameter 4 weeks after CaPO4 application. The maximum diameters of the aortas were measured by morphometry and the mean maximal diameters were calculated. Paraffin sections of the infrarenal aortas were examined for morphological analysis and immunohistochemical staining. The results showed that myeloid cell mPGES-1 deletion significantly mitigated AAA formation, including reducing expansion of the infrarenal aorta, preventing elastic lamellar degradation, and decreasing aortic calcium deposition. Immunohistochemical staining further indicated that macrophage infiltration and matrix metalloproteinase 2 (MMP2) expression was attenuated in the Mac-mPGES-1-KO aortas. Consistently, in vitro experiments showed that expression of pro-inflammatory cytokines and MMPs was significantly reduced when mPGES-1 was lacking in the primary cultured peritoneal macrophages. These data altogether demonstrated that deletion of mPGES-1 in myeloid cells may attenuate AAA formation and targeting myeloid cell mPGES-1 could potentially offer an effective strategy for the treatment and prevention of vascular inflammatory diseases.
Collapse
Affiliation(s)
- Meina Guo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Shuang Ji
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Hui Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Jiayang Zhang
- Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Jingwen Zhu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Guangrui Yang
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Lihong Chen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
42
|
Wang X, Wang M, Zhu TT, Zheng ZJ, Li S, Sui ZY, Guo X, Wu S, Zhang NN, Yu ZY, Hu CP, Tang YB, Wang Q, Zhang Z. The TRPM7 chanzyme in smooth muscle cells drives abdominal aortic aneurysm in mice. NATURE CARDIOVASCULAR RESEARCH 2025; 4:216-234. [PMID: 39953275 DOI: 10.1038/s44161-025-00613-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 01/15/2025] [Indexed: 02/17/2025]
Abstract
Ionic signaling in smooth muscle cells (SMCs) is critical for vascular homeostasis. In this study, we untangled the role of the bifunctional TRPM7 channel kinase (chanzyme) in abdominal aortic aneurysm (AAA) pathogenesis. Comparing SMC-specific, macrophage-specific and endothelial cell-specific Trpm7 knockout, we revealed that SMC-specific Trpm7 deficiency protected mice from AAA in two distinct preclinical models of the disease. We showed that the TRPM7 channel activity increased the Ca2+ and Zn2+ influx and the Ca2+/calcineurin/CRTC2/CREB-dependent and Zn2+/MTF1-dependent Mmp2 transcription. Repurposing the clinical drug FTY720 to prevent and treat AAA resulted in improved aortic phenotypes through inhibition of TRPM7 channel activity. This study highlights the ionic mechanisms underlying AAA, identifies TRPM7 as a potential therapeutic target and suggests that blocking TRPM7 channels could be a viable strategy for treating AAA.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/enzymology
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- TRPM Cation Channels/genetics
- TRPM Cation Channels/metabolism
- TRPM Cation Channels/deficiency
- TRPM Cation Channels/antagonists & inhibitors
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Mice, Knockout
- Aorta, Abdominal/enzymology
- Aorta, Abdominal/pathology
- Aorta, Abdominal/drug effects
- Male
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/deficiency
- Mice, Inbred C57BL
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 2/genetics
- Zinc/metabolism
- Calcium/metabolism
- Humans
- Mice
- Calcium Signaling/drug effects
- Cells, Cultured
Collapse
Affiliation(s)
- Xuan Wang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Mi Wang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Hunan Key Laboratory of Cardiometabolic Medicine, Central South University, Changsha, China
| | - Tian-Tian Zhu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zi-Jie Zheng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Shuang Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zhao-Yi Sui
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xin Guo
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sha Wu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Nai-Ning Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhi-Yi Yu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chang-Ping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, China
| | - Yong-Bo Tang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Qing Wang
- Department of Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, China.
| |
Collapse
|
43
|
Cai H, Li H, Xiao X, Wang S, Liu R, Qin Y, Zhou Y, Yao C. TRAF6 promotes abdominal aortic aneurysm development by activating macrophage pyroptosis via the NLRP3/Caspase1/GSDMD pathway. FASEB J 2025; 39:e70318. [PMID: 39831511 DOI: 10.1096/fj.202402873r] [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: 11/17/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
Abdominal aortic aneurysm represents a critical pathology of the aorta that currently lacks effective pharmacological interventions. TNF receptor-associated factor 6 (TRAF6) has been established to be involved in cardiovascular diseases such as atherosclerosis, hypertension, and heart failure. However, its role in abdominal aortic aneurysm (AAA) remains unclear. This study aimed to explore the role of TRAF6 on AAA formation and its underlying mechanisms. Single-cell RNA sequencing of human AAA tissues demonstrated that TRAF6 was significantly upregulated in aortic macrophages. Moreover, overexpression of TRAF6 promotes AAA formation in elastase-induced C57BL/6 mice, while TRAF6 pharmacological inhibition could attenuate AAA development. Consistently, inhibition of TRAF6 in macrophages through in vitro methods notably limits their pyroptosis, while also diminishing proinflammatory responses in these cells. Mechanistically, TRAF6 can modulate macrophage pyroptosis through the NLRP3/Caspase1/GSDMD signaling pathway. Our study highlights the crucial role of the TRAF6/NLRP3/Caspase1/GSDMD axis in macrophage pyroptosis and AAA, offering potential biomarkers and therapeutic targets for AAA.
Collapse
Affiliation(s)
- Huoying Cai
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Huaming Li
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaoyong Xiao
- Department of Emergency Medicine, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Siwen Wang
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Ruiming Liu
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Yuansen Qin
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Yu Zhou
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Chen Yao
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| |
Collapse
|
44
|
Sajjadi SM, Mohebbi A, Ehsani A, Marashi A, Azhdarimoghaddam A, Karami S, Karimi MA, Sadeghi M, Firoozi K, Mohammad Zamani A, Rigi A, Nayebagha M, Asadi Anar M, Eini P, Salehi S, Rostami Ghezeljeh M. Identifying abdominal aortic aneurysm size and presence using Natural Language Processing of radiology reports: a systematic review and meta-analysis. Abdom Radiol (NY) 2025:10.1007/s00261-025-04810-5. [PMID: 39883167 DOI: 10.1007/s00261-025-04810-5] [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: 10/25/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND AND AIM Prior investigations of the natural history of abdominal aortic aneurysms (AAAs) have been constrained by small sample sizes or uneven assessments of aggregated data. Natural language processing (NLP) can significantly enhance the investigation and treatment of patients with AAAs by swiftly and effectively collecting imaging data from health records. This meta-analysis aimed to evaluate the efficacy of NLP techniques in reliably identifying the existence or absence of AAAs and measuring the maximal abdominal aortic diameter in extensive datasets of radiology study reports. METHOD The PubMed, Scopus, Web of Science, Embase, and Science Direct databases were searched until March 2024 to obtain pertinent papers. The RAYYAN intelligent tool for systematic reviews was utilized to screen the studies. The meta-analysis was conducted using STATA v18 software. Egger's test was employed to evaluate publication bias. The Newcastle Ottawa Scale was employed to assess the quality of the listed studies. A plot digitizer was employed to extract digital data. RESULT A total of 39,094 individuals with AAA were included in this analysis. Twenty-seven thousand three hundred twenty-six patients were male, and 11,383 were female. The mean age of the total participants was 73.1 ± 1.25 years. Analysis results for pooled estimation of performance variables such as: The sensitivity, specificity, precision, and accuracy of the implemented NLP model were analyzed as follows: 0.89(0.88-0.91), 0.88 (0.87-0.89), 0.92 (0.89-0.95), and 0.91 (0.89-0.93) respectively. The aneurysm diameter size difference reported in follow-up before and after NLP implementation in the included studies showed a 0.05 cm reduction in size, which was statistically significant. CONCLUSION NLP holds great potential for automating the detection of AAA size and presence in radiology reports, enhancing efficiency and scalability over manual review. However, challenges persist. Variability in report formats, terminology, and unstructured data can compromise accuracy. Additionally, NLP models rely on high-quality, annotated training datasets, which may be incomplete or unrepresentative. While NLP aids in identifying AAA-related data, human oversight is essential to ensure decisions are informed by the patient's broader clinical context. Ongoing algorithm refinement and seamless integration into clinical workflows are key to improving NLP's utility and reliability in this field.
Collapse
Affiliation(s)
| | - Alisa Mohebbi
- Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | | | - Amir Marashi
- Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | | | - Shaghayegh Karami
- Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Mohammad Amin Karimi
- Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Mahsa Sadeghi
- Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Kiana Firoozi
- Gonabad University of Medical Sciences, Gonābād, Islamic Republic of Iran
| | - Amir Mohammad Zamani
- Ahvaz Jundishapur University of Medical Sciences, Ahvāz, Islamic Republic of Iran
| | - Amirhossein Rigi
- Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Melika Nayebagha
- Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | | | - Pooya Eini
- Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Sadaf Salehi
- Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | | |
Collapse
|
45
|
Hu K, Zhong L, Zhang J. Reply to "Comment on 'Pathogenesis-Guided Rational Engineering of Nanotherapies for the Targeted Treatment of Abdominal Aortic Aneurysm by Inhibiting Neutrophilic Inflammation'". ACS NANO 2025; 19:2977-2978. [PMID: 39871702 DOI: 10.1021/acsnano.4c18041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Affiliation(s)
- Kaiyao Hu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Ling Zhong
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
- Yu-Yue Pathology Scientific Research Center, 313 Gaoteng Avenue, Jiulongpo District, Chongqing 400039, P. R. China
| |
Collapse
|
46
|
Callow B, He X, Juriga N, Mangum KD, Joshi A, Xing X, Obi A, Chattopadhyay A, Milewicz DM, O’Riordan MX, Gudjonsson J, Gallagher K, Davis FM. Inhibition of vascular smooth muscle cell PERK/ATF4 ER stress signaling protects against abdominal aortic aneurysms. JCI Insight 2025; 10:e183959. [PMID: 39846252 PMCID: PMC11790032 DOI: 10.1172/jci.insight.183959] [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: 06/24/2024] [Accepted: 10/08/2024] [Indexed: 01/24/2025] Open
Abstract
Abdominal aortic aneurysms (AAA) are a life-threatening cardiovascular disease for which there is a lack of effective therapy preventing aortic rupture. During AAA formation, pathological vascular remodeling is driven by vascular smooth muscle cell (VSMC) dysfunction and apoptosis, for which the mechanisms regulating loss of VSMCs within the aortic wall remain poorly defined. Using single-cell RNA-Seq of human AAA tissues, we identified increased activation of the endoplasmic reticulum stress response pathway, PERK/eIF2α/ATF4, in aortic VSMCs resulting in upregulation of an apoptotic cellular response. Mechanistically, we reported that aberrant TNF-α activity within the aortic wall induces VSMC ATF4 activation through the PERK endoplasmic reticulum stress response, resulting in progressive apoptosis. In vivo targeted inhibition of the PERK pathway, with VSMC-specific genetic depletion (Eif2ak3fl/fl Myh11-CreERT2) or pharmacological inhibition in the elastase and angiotensin II-induced AAA model preserved VSMC function, decreased elastin fragmentation, attenuated VSMC apoptosis, and markedly reduced AAA expansion. Together, our findings suggest that cell-specific pharmacologic therapy targeting the PERK/eIF2α/ATF4 pathway in VSMCs may be an effective intervention to prevent AAA expansion.
Collapse
MESH Headings
- Activating Transcription Factor 4/metabolism
- Activating Transcription Factor 4/genetics
- eIF-2 Kinase/metabolism
- eIF-2 Kinase/genetics
- eIF-2 Kinase/antagonists & inhibitors
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Endoplasmic Reticulum Stress/drug effects
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Animals
- Humans
- Mice
- Signal Transduction/drug effects
- Apoptosis/drug effects
- Male
- Myocytes, Smooth Muscle/metabolism
- Disease Models, Animal
- Eukaryotic Initiation Factor-2/metabolism
- Angiotensin II
- Mice, Inbred C57BL
Collapse
Affiliation(s)
| | - Xiaobing He
- Section of Vascular Surgery, Department of Surgery, and
| | | | | | - Amrita Joshi
- Section of Vascular Surgery, Department of Surgery, and
| | - Xianying Xing
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrea Obi
- Section of Vascular Surgery, Department of Surgery, and
| | | | - Dianna M. Milewicz
- University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Mary X. O’Riordan
- Department Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Johann Gudjonsson
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Katherine Gallagher
- Section of Vascular Surgery, Department of Surgery, and
- Department Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
47
|
Wang X, Sun J, Chang N, Liu M, Zhang S. Association between non-alcoholic fatty liver disease and progression of abdominal aortic aneurysm: a multicenter study. BMC Med Imaging 2025; 25:24. [PMID: 39833711 PMCID: PMC11749205 DOI: 10.1186/s12880-025-01559-7] [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: 12/10/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND The purpose of our study was to investigate the association between non-alcoholic fatty liver disease (NAFLD) and abdominal aortic aneurysms (AAA) progression using non-enhanced computed tomography (CT) and CT angiography (CTA). METHODS Patients with AAA and age- and sex-matched healthy subjects who underwent abdominal CTA and non-enhanced CT examination between January 2015 and January 2023 from four hospitals were retrospectively analyzed. Patients with AAA were divided into progression (growth rate > 10 mL/year) and non-progression groups, as well as those with NAFLD and without NAFLD, based on abdominal CT results. The Kaplan-Meier and Cox regression were used to investigate the association between NAFLD and AAA progression. RESULTS A total of 151 patients with AAA (mean age: 69.1 ± 10.5 years old, 133 men) were included, among which 66 patients (43.7%) had NAFLD. During a median of 10.7 months (6.0-76.0 months), 57 patients (37.7%) had AAA progression. The prevalence of NAFLD was significantly higher in the AAA group compared to the control group (43.7% vs. 31.1%, p = 0.024). Multivariable regression analysis revealed that the NAFLD was independently associated with AAA progression (HR, 4.28; 95% CI, 2.20-8.31; p < 0.001). The area under curve of combined NAFLD and AAA maximal diameter was 0.857 for predicting AAA progression. CONCLUSIONS NAFLD on non-enhanced CT is an independent predictor of AAA progression. It can improve the diagnostic efficacy of predicting the progression of abdominal aortic aneurysms. CLINICAL TRIAL NUMBER Not applicable. This research is a retrospective analysis.
Collapse
Affiliation(s)
- Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324 Jingwu Road, Jinan, Shandong, 251200, China
| | - Jingxiang Sun
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, Shandong, 251200, China
- Postgraduate Department, Shandong First Medical University, Shandong Academy of Medical Sciences, No.6699 Qingdao Road, Jinan, Shandong, 250117, China
| | - Na Chang
- Department of Medical Technology, Jinan Nursing Vocational College, No. 3636 Gangxi Road, Jinan, Shandong, 250021, China
| | - Menghan Liu
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, Shandong, 251200, China
| | - Shuai Zhang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324 Jingwu Road, Jinan, Shandong, 251200, China.
| |
Collapse
|
48
|
Zhang L, Chen S, Ning M, Guo S, Wen D, Wang H, Sun Y, Yang G, Wang Y, Xue S. Tea Polyphenol-Derived Carbon Dots Alleviate Abdominal Aortic Aneurysm Progression by Mitigating Oxidative Stress and Ferroptosis. ACS APPLIED BIO MATERIALS 2025; 8:688-703. [PMID: 39737545 DOI: 10.1021/acsabm.4c01549] [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] [Indexed: 01/01/2025]
Abstract
Abdominal aortic aneurysm (AAA) is a cardiovascular disease with potentially fatal consequences, yet effective therapies to prevent its progression remain unavailable. Oxidative stress is associated with AAA development. Carbon dots have reactive oxygen species-scavenging activity, while green tea extract exhibits robust antioxidant properties. However, the potential of green tea derived carbon dots in mitigating AAA progression has not been fully elucidated. In this study, tea polyphenol carbon dots (TP-CDs) were synthesized via hydrothermal methods and characterized for their antioxidant properties. The antioxidant effects of TP-CDs were evaluated, and TP-CDs' impact on phenotypic transformation, oxidative stress, apoptosis and ferroptosis was investigated comprehensively in an Ang II-induced AAA model, employing techniques such as Western blotting, flow cytometry, and immunohistochemistry. The results revealed that TP-CDs effectively alleviated oxidative stress induced by Ang II stimulation, thereby inhibiting phenotypic transformation, apoptosis, and ferroptosis in vivo. Furthermore, treatment with TP-CDs significantly attenuated AAA progression in a mouse AAA model. Overall, these findings demonstrate that TP-CDs reduced reactive oxygen species levels in the microenvironment and alleviated the progression of AAA, offering a promising therapeutic strategy for this condition.
Collapse
Affiliation(s)
- Luzheng Zhang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Shuyang Chen
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Mengling Ning
- MOE Key Laboratory of Laser Life Science and SATCM Third Grade Laboratory of Chinese Medicine and Photonics Technology, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| | - Suxiang Guo
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Dezhong Wen
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Heng Wang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Yujin Sun
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Guangdong Yang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, OntarioP3B 2R9, Canada
| | - Yuehong Wang
- State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai 200127, P. R. China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| |
Collapse
|
49
|
Lei C, Zhou Q, Lv L, Liu D, Qian H. Inhibition of GPR4 Attenuates the Formation of Abdominal Aortic Aneurysm Through Inhibiting the SP-1/VEGF-A Signaling. J Biochem Mol Toxicol 2025; 39:e70118. [PMID: 39799555 DOI: 10.1002/jbt.70118] [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: 09/17/2024] [Revised: 11/30/2024] [Accepted: 12/21/2024] [Indexed: 01/15/2025]
Abstract
Abdominal aortic aneurysm (AAA) is a severe cardiovascular disease (CVD) that is partly attributable to endothelial dysfunction, inflammatory response, and angiogenesis. G protein-coupled receptor 4 (GPR4), a proton-sensitive G protein-coupled receptor that is abundantly expressed in vascular endothelial cells, has been associated with numerous physiological functions. Nevertheless, its potential involvement in the development of AAA remains unexplored. In this study, we examined the impact of GPR4 deletion on the development of AAA in ApoE-deficient mice. The mice were categorized into four distinct groups: the ApoE-/- with saline group, the ApoE-/-GPR4-/- with saline group, the ApoE-/- with Ang II group, and the ApoE-/-GPR4-/- with Ang II group. AAA were induced in the ApoE-/- mice through the perfusion of angiotensin II (Ang II). Notably, GPR4 was substantially elevated in the AAA tissues from both human subjects and experimental mice. The deletion of GPR4 substantially decreased the formation of Ang II-induced AAA, damages to elastin, and the expression of aortic inflammatory cytokines interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α), as well as vascular endothelial growth factor A/vascular endothelial growth factor receptor 2 (VEGF-A/VEGF-R2), in ApoE-/- mice. Human aortic endothelial cells (HAECs) were transfected with lenti-viral GPR4 shRNA and subsequently stimulated with Ang II. Our findings indicate that the knockout of GPR4 attenuated Ang II-induced angiogenic tube formation in HAECs by decreasing the expression of VEGF-A and VEGF-R2. Furthermore, GPR4 knockout also hindered the activation of specificity protein-1 (SP-1) by reducing its expression and transcriptional activity. Notably, the overexpression of SP-1 reversed the inhibitory effects of GPR4 knockout on angiogenic tube formation and the expression of VEGF-A/VEGF-R2. This suggests that the protective effects of GPR4 knockout are achieved through the inhibition of SP-1. In summary, the absence of GPR4 impeded AAA formation, indicating that GPR4 could potentially serve as a therapeutic target for AAA.
Collapse
MESH Headings
- Animals
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/prevention & control
- Mice
- Humans
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor A/genetics
- Signal Transduction
- Male
- Mice, Knockout
- Angiotensin II
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
Collapse
Affiliation(s)
- Chenggang Lei
- Department of Cardiothoracic Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| | - Qian Zhou
- Department of Cardiothoracic Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| | - Lizhen Lv
- Department of Cardiothoracic Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| | - Di Liu
- Department of Cardiothoracic Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| | - Haiyun Qian
- Department of Cardiothoracic Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province, China
| |
Collapse
|
50
|
Qin S, Zhu C, Chen C, Sheng Z, Cao Y. An emerging double‑edged sword role of ferroptosis in cardiovascular disease (Review). Int J Mol Med 2025; 55:16. [PMID: 39540363 PMCID: PMC11573318 DOI: 10.3892/ijmm.2024.5457] [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: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
The pathophysiology of cardiovascular disease (CVD) is complex and presents a serious threat to human health. Cardiomyocyte loss serves a pivotal role in both the onset and progression of CVD. Among various forms of programmed cell death, ferroptosis, along with apoptosis, autophagy and pyroptosis, is closely linked to the advancement of CVD. Ferroptosis, a mechanism of cell death, is driven by the buildup of oxidized lipids and excess iron. This pathway is modulated by lipid, amino acid and iron metabolism. Key characteristics of ferroptosis include disrupted iron homeostasis, increased peroxidation of polyunsaturated fatty acids due to reactive oxygen species, decreased glutathione levels and inactivation of glutathione peroxidase 4. Treatments targeting ferroptosis could potentially prevent or alleviate CVD by inhibiting the ferroptosis pathway. Ferroptosis is integral to the pathogenesis of several types of CVD and inhibiting its occurrence in cardiomyocytes could be a promising therapeutic strategy for the future treatment of CVD. The present review provided an in‑depth analysis of advancements in understanding the mechanisms underlying ferroptosis. The present manuscript summarized the interplay between ferroptosis and CVDs, highlighting its dual roles in these conditions. Additionally, potential therapeutic targets within the ferroptosis pathway were discussed, alongside the current limitations and future directions of these novel treatment strategies. The present review may offer novel insights into preventive and therapeutic approaches for CVDs.
Collapse
Affiliation(s)
- Sirun Qin
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Can Zhu
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Chenyang Chen
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhe Sheng
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yu Cao
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|