1
|
Wang X, Ma X, Chen S, Fan M, Jin C, Chen Y, Wang S, Wang Z, Meng F, Zhang C, Yang L. Harnessing m1A modification: a new frontier in cancer immunotherapy. Front Immunol 2024; 15:1517604. [PMID: 39687616 PMCID: PMC11647001 DOI: 10.3389/fimmu.2024.1517604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
N1-methyladenosine (m1A) modification is an epigenetic change that occurs on RNA molecules, regulated by a suite of enzymes including methyltransferases (writers), demethylases (erasers), and m1A-recognizing proteins (readers). This modification significantly impacts the function of RNA and various biological processes by affecting the structure, stability, translation, metabolism, and gene expression of RNA. Thereby, m1A modification is closely associated with the occurrence and progression of cancer. This review aims to explore the role of m1A modification in tumor immunity. m1A affects tumor immune responses by directly regulating immune cells and indirectly modulating tumor microenvironment. Besides, we also discuss the implications of m1A-mediated metabolic reprogramming and its nexus with immune checkpoint inhibitors, unveiling promising avenues for immunotherapeutic intervention. Additionally, the m1AScore, established based on the expression patterns of m1A modification, can be used to predict tumor prognosis and guide personalized therapy. Our review underscores the significance of m1A modification as a burgeoning frontier in cancer biology and immuno-oncology, with the potential to revolutionize cancer treatment strategies.
Collapse
Affiliation(s)
- Xinru Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiaoqing Ma
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Minyan Fan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chenying Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yushi Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Shaodong Wang
- Affiliated Nanjing Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Zhiying Wang
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Fei Meng
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chengwan Zhang
- Department of Central Laboratory, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu, China
| | - Lin Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Zhang W, Gonzalez L, Li X, Bai H, Li Z, Taniguchi R, Langford J, Ohashi Y, Thaxton C, Aoyagi Y, Yatsula B, Martin KA, Goodwin J, Tellides G, Long X, Shu C, Dardik A. Endothelial TGF-β Signaling Regulates Endothelial-Mesenchymal Transition During Arteriovenous Fistula Remodeling in Mice With Chronic Kidney Disease. Arterioscler Thromb Vasc Biol 2024; 44:2509-2526. [PMID: 39297205 PMCID: PMC11593991 DOI: 10.1161/atvbaha.124.320933] [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] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/10/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Arteriovenous fistulae (AVF) are the preferred vascular access for hemodialysis in patients with end-stage kidney disease. Chronic kidney disease (CKD) is associated with endothelial injury, impaired AVF maturation, and reduced patency, as well as utilization. Because CKD is characterized by multiple pathophysiological processes that induce endothelial-to-mesenchymal transition (EndMT), we hypothesized that CKD promotes EndMT during venous remodeling and that disruption of endothelial TGF (transforming growth factor)-β signaling inhibits EndMT to prevent AVF failure even in the end-stage kidney disease environment. METHODS The mouse 5/6 nephrectomy and aortocaval fistula models were used. CKD was created via 5/6 nephrectomy, with controls of no (0/6) or partial (3/6) nephrectomy in C57BL/6J mice. AVF were created in mice with knockdown of TGF-βR1/R2 (TGF-β receptors type 1/2) in either smooth muscle cells or endothelial cells. AVF diameters and patency were measured and confirmed by serial ultrasound examination. AVF, both murine and human, were examined using Western blot, histology, and immunofluorescence. Human and mouse endothelial cells were used for in vitro experiments. RESULTS CKD accelerates TGF-β activation and promotes EndMT that is associated with increased AVF wall thickness and reduced patency in mice. Inhibition of TGF-β signaling in both endothelial cells and smooth muscle cells decreased smooth muscle cell proliferation in the AVF wall, attenuated EndMT, and was associated with reduced wall thickness, increased outward remodeling, and improved AVF patency. Human AVF also showed increased TGF-β signaling and EndMT. CONCLUSIONS CKD promotes EndMT and reduces AVF patency. Inhibition of TGF-β signaling, especially disruption of endothelial cell-specific TGF-β signaling, attenuates EndMT and improves AVF patency in mouse AVF. Inhibition of EndMT may be a therapeutic approach of translational significance to improve AVF patency in human patients with CKD.
Collapse
MESH Headings
- Animals
- Arteriovenous Shunt, Surgical/adverse effects
- Signal Transduction
- Mice, Inbred C57BL
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/physiopathology
- Disease Models, Animal
- Transforming Growth Factor beta/metabolism
- Vascular Remodeling
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Humans
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Male
- Receptor, Transforming Growth Factor-beta Type II/metabolism
- Receptor, Transforming Growth Factor-beta Type II/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Cells, Cultured
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/genetics
- Vascular Patency
- Nephrectomy
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice
- Graft Occlusion, Vascular/metabolism
- Graft Occlusion, Vascular/etiology
- Graft Occlusion, Vascular/pathology
- Graft Occlusion, Vascular/physiopathology
- Graft Occlusion, Vascular/prevention & control
- Vena Cava, Inferior/metabolism
- Vena Cava, Inferior/pathology
- Vena Cava, Inferior/physiopathology
- Cell Proliferation
- Epithelial-Mesenchymal Transition
- Renal Dialysis
- Endothelial-Mesenchymal Transition
Collapse
Affiliation(s)
- Weichang Zhang
- State Key Laboratory of Cardiovascular Diseases, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China (W.Z., C.S.)
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China (W.Z., X. Li, C.S.)
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Xin Li
- State Key Laboratory of Cardiovascular Diseases, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China (W.Z., C.S.)
| | - Hualong Bai
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Zhuo Li
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
- Division of Vascular Surgery, The University of Tokyo, Japan (R.T., Y.O.)
| | - John Langford
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Yuichi Ohashi
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
- Division of Vascular Surgery, The University of Tokyo, Japan (R.T., Y.O.)
| | - Carly Thaxton
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Yukihiko Aoyagi
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Kathleen A. Martin
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
- Department of Medicine (Cardiovascular Medicine), Yale Cardiovascular Research Center (K.A.M.), Yale School of Medicine, New Haven, CT
| | - Julie Goodwin
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
- Department of Pediatrics (J.G.), Yale School of Medicine, New Haven, CT
| | - George Tellides
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
- Division of Cardiac Surgery, Department of Surgery (G.T.), Yale School of Medicine, New Haven, CT
- Surgical Service, VA Connecticut Healthcare Systems, West Haven, CT (G.T., A.D.)
| | - Xiaochun Long
- Vascular Biology Center, Medical College of Georgia at Augusta University (X. Long)
| | - Chang Shu
- State Key Laboratory of Cardiovascular Diseases, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China (W.Z., C.S.)
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China (W.Z., X. Li, C.S.)
| | - Alan Dardik
- Vascular Biology and Therapeutics Program (W.Z., L.G., H.B., Z.L., R.T., J.L., Y.O., C.T., A.Y., B.Y., K.A.M., J.G., G.T., A.D.), Yale School of Medicine, New Haven, CT
- Division of Vascular and Endovascular Surgery, Department of Surgery (A.D.), Yale School of Medicine, New Haven, CT
- Department of Cellular and Molecular Physiology (A.D.), Yale School of Medicine, New Haven, CT
- Surgical Service, VA Connecticut Healthcare Systems, West Haven, CT (G.T., A.D.)
| |
Collapse
|
3
|
Zhang Y, Jiang Y, Yu Y, Feng G, Zhao Z, Zhang W, Li S, Li Y, Yang Z, Yan X, Gao X, Chen ZJ, Zhao H, Zhao S. snRNA-seq of human ovaries reveals heat shock proteins are associated with obesity related cancer risk. J Transl Med 2024; 22:1063. [PMID: 39593105 PMCID: PMC11590508 DOI: 10.1186/s12967-024-05898-x] [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: 08/30/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Obesity significantly impacts female reproductive health and increases the risk of gynecological tumors. However, the specific transcriptional changes that occur in the ovarian microenvironment during obesity-induced stress and the relationship between obesity and ovarian cancer remain unclear. METHODS Our study investigated the single-cell landscape of the ovarian cortex in individuals with varying BMI levels by snRNA-seq, revealing weight-stage related cellular composition deviations and expression profile irregularities. RESULTS Using single-cell high-dimensional Weighted Gene Co-expression Network Analysis (hdWGCNA), we identified distinct obesity-related gene modules within various subpopulations of stroma cells and blood vascular endothelial cells. Notably, we observed a negative correlation between BMI and heat shock protein (HSP) family genes. Specifically, we found that HSPD1 might function as a potential regulator of ovarian carcinogenesis and progression under conditions of obesity, as supported by our co-analysis with data from three bulk RNA-seq ovarian cancer databases. Our findings suggested that lower expression of HSPD1 indicated a poorer prognosis for ovarian cancer. CONCLUSIONS Our study identified a cluster of genes in ovarian cells that are suppressed by obesity, including those belonging to HSP family genes. These findings provide valuable insights for investigating the link between obesity and ovarian diseases.
Collapse
Affiliation(s)
- Yuhan Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012, Shandong, China
| | - Yonghui Jiang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Yunhai Yu
- Department of Obstetrics and Gynecology, The Second Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Gengchen Feng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012, Shandong, China
| | - Zihe Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012, Shandong, China
| | - Weihan Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012, Shandong, China
| | - Shumin Li
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Yimeng Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012, Shandong, China
| | - Ziyi Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012, Shandong, China
| | - Xueqi Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012, Shandong, China
| | - Xueying Gao
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012, Shandong, China
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, 250012, Shandong, China
| | - Han Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012, Shandong, China.
| | - Shigang Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012, Shandong, China.
| |
Collapse
|
4
|
Estrada AC, Irons L, Tellides G, Humphrey JD. Multiscale computational model of aortic remodeling following postnatal disruption of TGFβ signaling. J Biomech 2024; 169:112152. [PMID: 38763809 PMCID: PMC11141772 DOI: 10.1016/j.jbiomech.2024.112152] [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: 12/06/2023] [Revised: 03/20/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
The healthy adult aorta is a remarkably resilient structure, able to resist relentless cardiac-induced and hemodynamic loads under normal conditions. Fundamental to such mechanical homeostasis is the mechano-sensitive cell signaling that controls gene products and thus the structural integrity of the wall. Mouse models have shown that smooth muscle cell-specific disruption of transforming growth factor-beta (TGFβ) signaling during postnatal development compromises this resiliency, rendering the aortic wall susceptible to aneurysm and dissection under normal mechanical loading. By contrast, disruption of such signaling in the adult aorta appears to introduce a vulnerability that remains hidden under normal loading, but manifests under increased loading as experienced during hypertension. We present a multiscale (transcript to tissue) computational model to examine possible reasons for compromised mechanical homeostasis in the adult aorta following reduced TGFβ signaling in smooth muscle cells.
Collapse
Affiliation(s)
- Ana C Estrada
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Linda Irons
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - George Tellides
- Department of Surgery, Cardiothoracic, Yale School of Medicine, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
5
|
Jiang B, Ren P, He C, Wang M, Murtada SI, Chen Y, Ramachandra AB, Li G, Qin L, Assi R, Schwartz MA, Humphrey JD, Tellides G. Short-Term Disruption of TGFβ Signaling in Adult Mice Renders the Aorta Vulnerable to Hypertension-Induced Dissection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590484. [PMID: 38712205 PMCID: PMC11071440 DOI: 10.1101/2024.04.22.590484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Hypertension and transient increases in blood pressure from extreme exertion are risk factors for aortic dissection in patients with age-related vascular degeneration or inherited connective tissue disorders. Yet, the common experimental model of angiotensin II-induced aortopathy in mice appears independent of high blood pressure as lesions do not occur in response to an alternative vasoconstrictor, norepinephrine, and are not prevented by co-treatment with a vasodilator, hydralazine. We investigated vasoconstrictor administration to adult mice 1 week after disruption of TGFβ signaling in smooth muscle cells. Norepinephrine increased blood pressure and induced aortic dissection by 7 days and even within 30 minutes that was rescued by hydralazine; results were similar with angiotensin II. Changes in regulatory contractile molecule expression were not of pathological significance. Rather, reduced synthesis of extracellular matrix yielded a vulnerable aortic phenotype by decreasing medial collagen, most dynamically type XVIII, and impairing cell-matrix adhesion. We conclude that transient and sustained increases in blood pressure cause dissection in aortas rendered vulnerable by inhibition of TGFβ-driven extracellular matrix production by smooth muscle cells. A corollary is that medial fibrosis, a frequent feature of medial degeneration, may afford some protection against aortic dissection.
Collapse
|
6
|
Rego BV, Weiss D, Humphrey JD. A Fast, Robust Method for Quantitative Assessment of Collagen Fibril Architecture from Transmission Electron Micrographs. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:2099-2107. [PMID: 37856696 PMCID: PMC11419845 DOI: 10.1093/micmic/ozad116] [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: 05/03/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
Collagen is the most abundant protein in mammals; it exhibits a hierarchical organization and provides structural support to a wide range of soft tissues, including blood vessels. The architecture of collagen fibrils dictates vascular stiffness and strength, and changes therein can contribute to disease progression. While transmission electron microscopy (TEM) is routinely used to examine collagen fibrils under normal and pathological conditions, computational tools that enable fast and minimally subjective quantitative assessment remain lacking. In the present study, we describe a novel semi-automated image processing and statistical modeling pipeline for segmenting individual collagen fibrils from TEM images and quantifying key metrics of interest, including fibril cross-sectional area and aspect ratio. For validation, we show first-of-their-kind illustrative results for adventitial collagen in the thoracic aorta from three different mouse models.
Collapse
Affiliation(s)
- Bruno V. Rego
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Biological & Agricultural Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - Dar Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
7
|
Weiss D, Rego BV, Cavinato C, Li DS, Kawamura Y, Emuna N, Humphrey JD. Effects of Age, Sex, and Extracellular Matrix Integrity on Aortic Dilatation and Rupture in a Mouse Model of Marfan Syndrome. Arterioscler Thromb Vasc Biol 2023; 43:e358-e372. [PMID: 37470181 PMCID: PMC10528515 DOI: 10.1161/atvbaha.123.319122] [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] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Transmural failure of the aorta is responsible for substantial morbidity and mortality; it occurs when mechanical stress exceeds strength. The aortic root and ascending aorta are susceptible to dissection and rupture in Marfan syndrome, a connective tissue disorder characterized by a progressive reduction in elastic fiber integrity. Whereas competent elastic fibers endow the aorta with compliance and resilience, cross-linked collagen fibers confer stiffness and strength. We hypothesized that postnatal reductions in matrix cross-linking increase aortopathy when turnover rates are high. METHODS We combined ex vivo biaxial mechanical testing with multimodality histological examinations to quantify expected age- and sex-dependent structural vulnerability of the ascending aorta in Fbn1C1041G/+ Marfan versus wild-type mice without and with 4-week exposures to β-aminopropionitrile, an inhibitor of lysyl oxidase-mediated cross-linking of newly synthesized elastic and collagen fibers. RESULTS We found a strong β-aminopropionitrile-associated sexual dimorphism in aortic dilatation in Marfan mice and aortic rupture in wild-type mice, with dilatation correlating with compromised elastic fiber integrity and rupture correlating with compromised collagen fibril organization. A lower incidence of rupture of β-aminopropionitrile-exposed Marfan aortas associated with increased lysyl oxidase, suggesting a compensatory remodeling of collagen that slows disease progression in the otherwise compromised Fbn1C1041G/+ aorta. CONCLUSIONS Collagen fiber structure and function in the Marfan aorta are augmented, in part, by increased lysyl oxidase in female and especially male mice, which improves structural integrity, particularly via fibrils in the adventitia. Preserving or promoting collagen cross-linking may represent a therapeutic target for an otherwise vulnerable aorta.
Collapse
Affiliation(s)
- Dar Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - Bruno V Rego
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - David S Li
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - Yuki Kawamura
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - Nir Emuna
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.V.R., C.C., D.S.L., Y.K., N.E., J.D.H.)
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.)
| |
Collapse
|
8
|
Anderl WJ, Pearson N, Converse MI, Yu SM, Monson KL. Strain-induced collagen denaturation is rate dependent in failure of cerebral arteries. Acta Biomater 2023; 164:282-292. [PMID: 37116635 DOI: 10.1016/j.actbio.2023.04.032] [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: 10/29/2022] [Revised: 04/06/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023]
Abstract
While soft tissues are commonly damaged by mechanical loading, the manifestation of this damage at the microstructural level is not fully understood. Specifically, while rate-induced stiffening has been previously observed in cerebral arteries, associated changes in microstructural damage patterns following high-rate loading are largely undefined. In this study, we stretched porcine middle cerebral arteries to failure at 0.01 and >150 s-1, both axially and circumferentially, followed by probing for denatured tropocollagen using collagen hybridizing peptide (CHP). We found that collagen fibrils aligned with the loading direction experienced less denaturation following failure tests at high than low rates. Others have demonstrated similar rate dependence in tropocollagen denaturation during soft tissue failure, but this is the first study to quantify this behavior using CHP and to report it for cerebral arteries. These findings may have significant implications for traumatic brain injury and intracranial balloon angioplasty. We additionally observed possible tropocollagen denaturation in vessel layers primarily composed of fibrils transversely aligned to the loading axis. To our knowledge, this is the first observation of collagen denaturation due to transverse loading, but further research is needed to confirm this finding. STATEMENT OF SIGNIFICANCE: Previous work shows that collagen hybridizing peptide (CHP) can be used to identify collagen molecule unfolding and denaturation in mechanically overloaded soft tissues, including the cerebral arteries. But experiments have not explored collagen damage at rates relevant to traumatic brain injury. In this work, we quantified collagen damage in cerebral arteries stretched to failure at both high and low rates. We found that the collagen molecule is less damaged at high than at low rates, suggesting that damage mechanisms of either the collagen molecule or other elements of the collagen superstructure are rate dependent. This work implies that arteries failed at high rates, such as in traumatic brain injury, will have different molecular-level damage patterns than arteries failed at low rates. Consequently, improved understanding of damage characteristics may be expanded in the future to better inform clinically relevant cases of collagen damage such as angioplasty and injury healing.
Collapse
Affiliation(s)
| | - Noah Pearson
- DepSSSartment of Mechanical Engineering, University of Utah
| | | | - S Michael Yu
- Department of Biomedical Engineering, University of Utah; Department of Molecular Pharmaceutics, University of Utah
| | - Kenneth L Monson
- DepSSSartment of Mechanical Engineering, University of Utah; Department of Biomedical Engineering, University of Utah.
| |
Collapse
|
9
|
Salarian M, Ghim M, Toczek J, Han J, Weiss D, Spronck B, Ramachandra AB, Jung JJ, Kukreja G, Zhang J, Lakheram D, Kim SK, Humphrey JD, Sadeghi MM. Homeostatic, Non-Canonical Role of Macrophage Elastase in Vascular Integrity. Circ Res 2023; 132:432-448. [PMID: 36691905 PMCID: PMC9930896 DOI: 10.1161/circresaha.122.322096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Matrix metalloproteinase (MMP)-12 is highly expressed in abdominal aortic aneurysms and its elastolytic function has been implicated in the pathogenesis. This concept is challenged, however, by conflicting data. Here, we sought to revisit the role of MMP-12 in abdominal aortic aneurysm. METHODS Apoe-/- and Mmp12-/-/Apoe-/- mice were infused with Ang II (angiotensin). Expression of neutrophil extracellular traps (NETs) markers and complement component 3 (C3) levels were evaluated by immunostaining in aortas of surviving animals. Plasma complement components were analyzed by immunoassay. The effects of a complement inhibitor, IgG-FH1-5 (factor H-immunoglobulin G), and macrophage-specific MMP-12 deficiency on adverse aortic remodeling and death from rupture in Ang II-infused mice were determined. RESULTS Unexpectedly, death from aortic rupture was significantly higher in Mmp12-/-/Apoe-/- mice. This associated with more neutrophils, citrullinated histone H3 and neutrophil elastase, markers of NETs, and C3 levels in Mmp12-/- aortas. These findings were recapitulated in additional models of abdominal aortic aneurysm. MMP-12 deficiency also led to more pronounced elastic laminae degradation and reduced collagen integrity. Higher plasma C5a in Mmp12-/- mice pointed to complement overactivation. Treatment with IgG-FH1-5 decreased aortic wall NETosis and reduced adverse aortic remodeling and death from rupture in Ang II-infused Mmp12-/- mice. Finally, macrophage-specific MMP-12 deficiency recapitulated the effects of global MMP-12 deficiency on complement deposition and NETosis, as well as adverse aortic remodeling and death from rupture in Ang II-infused mice. CONCLUSIONS An MMP-12 deficiency/complement activation/NETosis pathway compromises aortic integrity, which predisposes to adverse vascular remodeling and abdominal aortic aneurysm rupture. Considering these new findings, the role of macrophage MMP-12 in vascular homeostasis demands re-evaluation of MMP-12 function in diverse settings.
Collapse
Affiliation(s)
- Mani Salarian
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Mean Ghim
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Jakub Toczek
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Jinah Han
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Dar Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.S., A.B.R., J.D.H.)
| | - Bart Spronck
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.S., A.B.R., J.D.H.)
| | - Abhay B. Ramachandra
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.S., A.B.R., J.D.H.)
| | - Jae-Joon Jung
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Gunjan Kukreja
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Jiasheng Zhang
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | | | - Sung-Kwon Kim
- Alexion Pharmaceuticals, New Haven, CT (D.L., S.-K.K.)
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.S., A.B.R., J.D.H.)
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.)
| | - Mehran M. Sadeghi
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| |
Collapse
|
10
|
Rego BV, Weiss D, Humphrey JD. A fast, robust method for quantitative assessment of collagen fibril architecture from transmission electron micrographs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527383. [PMID: 36798181 PMCID: PMC9934578 DOI: 10.1101/2023.02.06.527383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Collagen is the most abundant protein in mammals; it exhibits a hierarchical organization and provides structural support to a wide range of soft tissues, including blood vessels. The architecture of collagen fibrils dictates vascular stiffness and strength, and changes therein can contribute to disease progression. While transmission electron microscopy (TEM) is routinely used to examine collagen fibrils under normal and pathological conditions, computational tools that enable fast and minimally subjective quantitative assessment remain lacking. In the present study, we describe a novel semi-automated image processing and statistical modeling pipeline for segmenting individual collagen fibrils from TEM images and quantifying key metrics of interest, including fibril cross-sectional area and aspect ratio. For validation, we show illustrative results for adventitial collagen in the thoracic aorta from three different mouse models.
Collapse
Affiliation(s)
- Bruno V. Rego
- Department of Biomedical Engineering, School of Engineering & Applied Science, Yale University, New Haven, CT, USA
| | - Dar Weiss
- Department of Biomedical Engineering, School of Engineering & Applied Science, Yale University, New Haven, CT, USA
| | - Jay D. Humphrey
- Department of Biomedical Engineering, School of Engineering & Applied Science, Yale University, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
11
|
Linka K, Cavinato C, Humphrey JD, Cyron CJ. Predicting and understanding arterial elasticity from key microstructural features by bidirectional deep learning. Acta Biomater 2022; 147:63-72. [PMID: 35643194 DOI: 10.1016/j.actbio.2022.05.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 01/15/2023]
Abstract
Microstructural features and mechanical properties are closely related in all soft biological tissues. Both yet exhibit considerable inter-individual differences and are affected by factors such as aging and disease and its progression. Histological analysis, modern in situ imaging, and biomechanical testing have deepened our understanding of these complex interrelations, yet two key questions remain: (1) Given the specific microstructure, can one predict the macroscopic mechanical properties without mechanical testing? (2) Can one quantify individual contributions of the different microstructural features to the macroscopic mechanical properties in an automated, systematic and largely unbiased way? Here we propose a bidirectional deep learning architecture to address these two questions. Our architecture uses data from standard histological analyses, two-photon microscopy and biaxial biomechanical testing. Its capabilities are demonstrated by predicting with high accuracy (R2=0.92) the evolving mechanical properties of the murine aorta during maturation and aging. Moreover, our architecture reveals that the extracellular matrix composition and organization are the most prominent factors governing the macroscopic mechanical properties of the tissues studied herein. STATEMENT OF SIGNIFICANCE: .
Collapse
Affiliation(s)
- Kevin Linka
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Hamburg, Germany
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Christian J Cyron
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Hamburg, Germany; Institute of Material Systems Modeling, Helmholtz-Zentrum Hereon, Geesthacht, Germany.
| |
Collapse
|
12
|
Critical Pressure of Intramural Delamination in Aortic Dissection. Ann Biomed Eng 2022; 50:183-194. [PMID: 35044571 PMCID: PMC8957392 DOI: 10.1007/s10439-022-02906-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 01/01/2022] [Indexed: 02/03/2023]
Abstract
Computational models of aortic dissection can examine mechanisms by which this potentially lethal condition develops and propagates. We present results from phase-field finite element simulations that are motivated by a classical but seldom repeated experiment. Initial simulations agreed qualitatively and quantitatively with data, yet because of the complexity of the problem it was difficult to discern trends. Simplified analytical models were used to gain further insight. Together, simplified and phase-field models reveal power-law-based relationships between the pressure that initiates an intramural tear and key geometric and mechanical factors-insult surface area, wall stiffness, and tearing energy. The degree of axial stretch and luminal pressure similarly influence the pressure of tearing, which was ~88 kPa for healthy and diseased human aortas having sub-millimeter-sized initial insults, but lower for larger tear sizes. Finally, simulations show that the direction a tear propagates is influenced by focal regions of weakening or strengthening, which can drive the tear towards the lumen (dissection) or adventitia (rupture). Additional data on human aortas having different predisposing disease conditions will be needed to extend these results further, but the present findings show that physiologic pressures can propagate initial medial defects into delaminations that can serve as precursors to dissection.
Collapse
|
13
|
Estrada AC, Irons L, Rego BV, Li G, Tellides G, Humphrey JD. Roles of mTOR in thoracic aortopathy understood by complex intracellular signaling interactions. PLoS Comput Biol 2021; 17:e1009683. [PMID: 34898595 PMCID: PMC8700007 DOI: 10.1371/journal.pcbi.1009683] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/23/2021] [Accepted: 11/26/2021] [Indexed: 02/01/2023] Open
Abstract
Thoracic aortopathy–aneurysm, dissection, and rupture–is increasingly responsible for significant morbidity and mortality. Advances in medical genetics and imaging have improved diagnosis and thus enabled earlier prophylactic surgical intervention in many cases. There remains a pressing need, however, to understand better the underlying molecular and cellular mechanisms with the hope of finding robust pharmacotherapies. Diverse studies in patients and mouse models of aortopathy have revealed critical changes in multiple smooth muscle cell signaling pathways that associate with disease, yet integrating information across studies and models has remained challenging. We present a new quantitative network model that includes many of the key smooth muscle cell signaling pathways and validate the model using a detailed data set that focuses on hyperactivation of the mechanistic target of rapamycin (mTOR) pathway and its inhibition using rapamycin. We show that the model can be parameterized to capture the primary experimental findings both qualitatively and quantitatively. We further show that simulating a population of cells by varying receptor reaction weights leads to distinct proteomic clusters within the population, and that these clusters emerge due to a bistable switch driven by positive feedback in the PI3K/AKT/mTOR signaling pathway. Cell signaling drives changes across scales, from altered transcription at the single-cell level to tissue-level growth and remodeling. Studying complex interactions within cell signaling pathways can lead to a better understanding of the progression of disease. In particular, we are interested in how vascular cells can change their phenotype in a way that exacerbates aortopathy, namely, the development of aneurysms, dissections, and rupture. In this study we built a novel cell signaling network model of a vascular smooth muscle cell using archival data and used it to capture the effects of a genetic knock-out and subsequent pharmacologic rescue. We then used the model to simulate populations of smooth muscle cells and found that small perturbations to the strength of signaling can lead to distinct clusters of cells. With further analysis of the network substructures, we found that a positive feedback loop within the network was responsible for the distinct phenotypes we saw in our clusters of simulated cells. We believe that this work not only helps us to understand changes in smooth muscle cell phenotype but also opens the possibility to study other signaling perturbations associated with aortopathy.
Collapse
Affiliation(s)
- Ana C. Estrada
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
| | - Linda Irons
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
| | - Bruno V. Rego
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
| | - Guangxin Li
- Department of Surgery, Yale School of Medicine; New Haven, Connecticut, United States of America
| | - George Tellides
- Department of Surgery, Yale School of Medicine; New Haven, Connecticut, United States of America
- Vascular Biology and Therapeutics Program, Yale School of Medicine; New Haven, Connecticut, United States of America
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
- Vascular Biology and Therapeutics Program, Yale School of Medicine; New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
14
|
Nicotine Exacerbates TAAD Formation Induced by Smooth Muscle-Specific Deletion of the TGF- β Receptor 2. J Immunol Res 2021; 2021:6880036. [PMID: 34646889 PMCID: PMC8505064 DOI: 10.1155/2021/6880036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/04/2021] [Indexed: 01/22/2023] Open
Abstract
Tobacco smoke is an established risk factor for thoracic aortic aneurysms and dissections (TAAD). However, little is known about its underlying mechanisms due to the lack of validated animal models. The present study developed a mouse model that may be utilized to investigate exacerbation of TAAD formation by mimetics of tobacco smoke. TAADs were created via inducible deletion of smooth muscle cell-specific Tgfbr2 receptors. Using this model, the first set of experiments evaluated the efficacy of nicotine salt (34.0 mg/kg/day), nicotine free base (NFB, 5.0 mg 90-day pellets), and cigarette smoke extract (0.1 ml/mouse/day). Compared with their respective control groups, only NFB pellets promoted TAAD dilation (23 ± 3% vs. 12 ± 2%, P = 0.014), and this efficacy was achieved at a cost of >50% acute mortality. Infusion of NFB with osmotic minipumps at extremely high, but nonlethal, doses (15.0 or 45.0 mg/kg/day) failed to accelerate TAAD dilation. Interestingly, costimulation with β-aminopropionitrile (BAPN) promoted TAAD dilation and aortic rupture at dosages of 3.0 and 45.0 mg/kg/day, respectively, indicating that BAPN sensitizes the response of TAADs to NFB. In subsequent analyses, the detrimental effects of NFB were associated with clustering of macrophages, neutrophils, and T-cells in areas with structural destruction, enhanced matrix metalloproteinase- (MMP-) 2 production, and pathological angiogenesis with attenuated fibrosis in the adventitia. In conclusion, modeling nicotine exacerbation of TAAD formation requires optimization of chemical form, route of delivery, and dosage of the drug as well as the pathologic complexity of TAADs. Under the optimized conditions of the present study, chronic inflammation and adventitial mal-remodeling serve as critical pathways through which NFB exacerbates TAAD formation.
Collapse
|