1
|
Li M, Hu X, Hu X, Gao F, Cui Y, Wei X, Qin Y, An X, Zhao Y, Gao Y. Inhibition of PHB1/PHB2 suppresses atherosclerotic plaque formation by interrupting PI3K/AKT/mTOR signaling. PLoS One 2025; 20:e0320509. [PMID: 40168274 PMCID: PMC11960914 DOI: 10.1371/journal.pone.0320509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/19/2025] [Indexed: 04/03/2025] Open
Abstract
Prohibitin 1 (PHB1) and prohibitin 2 (PHB2) are highly conserved proteins belonging to the stomatin-prohibitin flotillin-HflC/K (SPFH) protein superfamily. They are ubiquitously expressed and implicated in the regulation of cell proliferation, migration, and survival. However, the expression and biological functions of PHB1/PHB2 in atherosclerosis (AS) remain unclear. In the present study, an enzyme-linked immunosorbent assay was used to detect PHB1/PHB2 expression in the serum of patients with hyperlipidemia. The potential effect and mechanism of PHB1/PHB2 in apolipoprotein E-deficient (ApoE-/-) mice were also investigated. shRNA-PHB1 and shRNA-PHB2 lentiviruses were engineered and tail vein-injected into ApoE-/- mice fed a high-fat diet. IL-8, a proatherogenic cytokine, was used as an inducer in vitro. The effects of a PHB1/PHB2 knockdown on vascular smooth muscle cell (VSMC) proliferation, migration, and autophagy and endothelial cell (EC) adhesion were evaluated using methyl thiazolyl tetrazolium (MTT), Transwell migration, Boyden chamber, and monocyte adhesion assays, as well as transmission electron microscopy. Compared with the healthy subjects, PHB1/PHB2 expression was elevated in the serum of patients with hyperlipidemia. Animal experiments showed that downregulation of PHBs reduced the area of atherosclerotic lesions, and the expression of cyclinD1, MMP9, and LC3. In addition, in vitro experiments showed that downregulating PHB1/PHB2 expression under inflammatory stimulation reduced the adhesion, proliferation, migration, and autophagy of ECs and VSMCs by inhibiting the PI3K/Akt/mTOR pathway activation. Collectively, our findings showed that PHBs are activly associated with AS progression.
Collapse
Affiliation(s)
- Mei Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiaoyan Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xinxin Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Fuhua Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Cui
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Xiaoqing Wei
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Yuanhua Qin
- Department of Parasite, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaohua An
- Clinical Laboratory, Dalian Central Hospital, Dalian, China
| | - Ying Zhao
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Ying Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| |
Collapse
|
2
|
Parveen K, Hussain MA, Anwar S, Elagib HM, Kausar MA. Comprehensive review on diabetic foot ulcers and neuropathy: Treatment, prevention and management. World J Diabetes 2025; 16:100329. [PMID: 40093290 PMCID: PMC11885961 DOI: 10.4239/wjd.v16.i3.100329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/27/2024] [Accepted: 12/27/2024] [Indexed: 01/21/2025] Open
Abstract
Diabetic foot (DF) is a major public health concern. As evident from numerous previous studies, supervision of DF ulcer (DFU) is crucial, and a specific quality check-up is needed. Patients should be educated about glycaemic management, DFUs, foot lesions, proper care for injuries, diet, and surgery. Certain reasonably priced treatments, such as hyperbaric oxygen and vacuum-assisted closure therapy, are also available for DFUs, along with modern wound care products and techniques. Nonetheless, DF care (cleaning, applying antimicrobial cream when wounded, and foot reflexology), blood glucose monitoring to control diabetes, and monthly or quarterly examinations in individuals with diabetes are effective in managing DFUs. Between 50% and 80% of DF infections are preventable. Regardless of the intensity of the lesion, it needs to be treated carefully and checked daily during infection. Tissue regeneration can be aided by cleaning, dressing, and application of topical medicines. The choice of shoes is also important because it affects blood circulation and nerve impulses. In general, regular check-ups, monitoring of the patient's condition, measuring blood glucose levels, and providing frequent guidance regarding DFU care are crucial. Finally, this important clinical problem requires involvement of multiple professionals to properly manage it.
Collapse
Affiliation(s)
- Kehkashan Parveen
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Malik Asif Hussain
- Department of Pathology, College of Medicine, University of Ha’il, Ha'il 53962, Saudi Arabia
| | - Sadaf Anwar
- Department of Biochemistry, College of Medicine, University of Ha’il, Ha'il 53962, Saudi Arabia
| | | | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Ha’il, Ha'il 53962, Saudi Arabia
| |
Collapse
|
3
|
Li T, Lin S, Zhu Y, Ye D, Rong X, Wang L. Basic biology and roles of CEBPD in cardiovascular disease. Cell Death Discov 2025; 11:102. [PMID: 40087290 PMCID: PMC11909146 DOI: 10.1038/s41420-025-02357-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/25/2025] [Accepted: 02/12/2025] [Indexed: 03/17/2025] Open
Abstract
CCAAT/enhancer-binding protein delta (CEBPD), as an evolutionarily conserved protein in mammals, belongs to the CEBP transcription factor family, which modulates many biological processes. The diversity of CEBPD functions partly depends on the cell type and cellular context. Aberrant CEBPD expression and activity are associated with multiple organ diseases, including cardiovascular diseases. In this review, we describe the basic molecular biology of CEBPD to understand its expression regulation, modifications, and functions. Here, we summarize the recent advances in genetically modified animals with CEBPD. Finally, we discuss the contribution of CEBPD to cardiovascular diseases and highlight the strategies for developing novel therapies targeting CEBPD.
Collapse
Affiliation(s)
- Tongjun Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Shaoling Lin
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Yingyin Zhu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Dewei Ye
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China.
| | - Xianglu Rong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China.
| | - Lexun Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
4
|
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
|
5
|
Liu Y, Zuo M, Wu A, Wang Z, Wang S, Bai Y, Zhou J, Wang H. UFMylation maintains YAP stability to promote vascular endothelial cell senescence. iScience 2025; 28:111854. [PMID: 39991547 PMCID: PMC11847039 DOI: 10.1016/j.isci.2025.111854] [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: 07/24/2024] [Revised: 10/30/2024] [Accepted: 01/17/2025] [Indexed: 02/25/2025] Open
Abstract
Endothelial cell (EC) senescence is an accomplice for vascular aging, which leads to cardiovascular diseases (CVDs). Evidences showed that Hippo-Yes-associated protein (YAP) signaling pathway plays an essential role in aging-associated CVDs. Here, we reported that YAP was elevated in senescent human umbilical vein endothelial cells (HUVECs) and inhibition of YAP could attenuate HUVECs senescence. Besides, our findings revealed that the activity of UFMylation and the level of YAP were both elevated in senescent cells. Furthermore, UFM1-modified YAP was upregulated in senescent ECs, and increased the stability of YAP. Importantly, we found that compound 8.5, an inhibitor of E1 of UFMylation, can alleviate vascular aging in aged mice. Together, our finding provides molecular mechanism by which UFMylation maintains YAP stability and exerts an important role in promoting cell senescence, and identified that a previously unrecognized UFMylation is a potential therapeutic target for anti-aging.
Collapse
Affiliation(s)
- Yanan Liu
- Department of Geriatric Medicine, Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Institute of Aging Research, School of Basic Medicine Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Min Zuo
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Institute of Aging Research, School of Basic Medicine Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Aiwei Wu
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Institute of Aging Research, School of Basic Medicine Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Zhaoxiang Wang
- School of Basic Medicine, Guangdong Medical University, Dongguan 523808, China
| | - Siting Wang
- Department of Geriatric Medicine, Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yongping Bai
- Department of Geriatric Medicine, Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Junzhi Zhou
- School of Basic Medicine, Guangdong Medical University, Dongguan 523808, China
| | - Hu Wang
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Institute of Aging Research, School of Basic Medicine Sciences, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
6
|
Su C, Mi X, Ito T, Kato Y, Nishimura A, Nagata R, Mori Y, Nishida M. TRPC6-Mediated Zn 2+ Influx Negatively Regulates Contractile Differentiation of Vascular Smooth Muscle Cells. Biomolecules 2025; 15:267. [PMID: 40001570 PMCID: PMC11853136 DOI: 10.3390/biom15020267] [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: 12/27/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Vascular smooth muscle cells (VSMCs) can dynamically change their phenotype between contractile and synthetic forms in response to environmental stress, which is pivotal in maintaining vascular homeostasis and mediating pathological remodeling of blood vessels. We previously reported that suppression of canonical transient receptor potential 6 (TRPC6) channel-mediated cation entry sustains VSMCs contractile phenotype and promotes the blood flow recovery after hindlimb ischemia in mice. We also reported that Zn2+, a metal biomolecule mobilized by TRPC6 channel activation, exerts potential beneficial effects on cardiac contractility and remodeling. Therefore, we hypothesized that TRPC6-mediated Zn2+ influx participates in phenotype switching of VSMCs and vascular remodeling. We established rat aortic smooth muscle cells (RAoSMCs) stably expressing wild type (WT) and Zn2+ only impermeable TRPC6 (KYD) mutant. Although the resting phenotypes were similar in both RAoSMCs, pharmacological TRPC6 activation by PPZ2 prevented the transforming growth factor (TGF) β-induced reduction in the intracellular Zn2+ amount and contractile differentiation in RAoSMCs (WT), but failed to prevent them in RAoSMCs (KYD). There were no significant differences in TRPC6-dependent cation currents among all RAoSMCs pretreated with or without TGFβ and/or PPZ2, suggesting that TRPC6 channels are functionally expressed in RAoSMCs regardless of their phenotype. Treatment of mice with PPZ2 attenuated the progression of vascular remodeling caused by chronic angiotensin II infusion. These results suggest that Zn2+ influx through TRPC6 channels negatively regulates the TGFβ-induced contractile differentiation of VSMCs and the progression of vascular remodeling in rodents.
Collapse
Affiliation(s)
- Chenlin Su
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan (Y.K.)
| | - Xinya Mi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan (Y.K.)
| | - Tomoya Ito
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan (Y.K.)
| | - Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan (Y.K.)
| | - Akiyuki Nishimura
- National Institute for Physiological Science (NIPS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Science, Okazaki 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| | - Ryu Nagata
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Yasuo Mori
- Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Motohiro Nishida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan (Y.K.)
- National Institute for Physiological Science (NIPS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Science, Okazaki 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| |
Collapse
|
7
|
Jiang M, Fang H, Tian H. Metabolism of cancer cells and immune cells in the initiation, progression, and metastasis of cancer. Theranostics 2025; 15:155-188. [PMID: 39744225 PMCID: PMC11667227 DOI: 10.7150/thno.103376] [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: 09/07/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025] Open
Abstract
The metabolism of cancer and immune cells plays a crucial role in the initiation, progression, and metastasis of cancer. Cancer cells often undergo metabolic reprogramming to sustain their rapid growth and proliferation, along with meeting their energy demands and biosynthetic needs. Nevertheless, immune cells execute their immune response functions through the specific metabolic pathways, either to recognize, attack, and eliminate cancer cells or to promote the growth or metastasis of cancer cells. The alteration of cancer niches will impact the metabolism of both cancer and immune cells, modulating the survival and proliferation of cancer cells, and the activation and efficacy of immune cells. This review systematically describes the key characteristics of cancer cell metabolism and elucidates how such metabolic traits influence the metabolic behavior of immune cells. Moreover, this article also highlights the crucial role of immune cell metabolism in anti-tumor immune responses, particularly in priming T cell activation and function. By comprehensively exploring the metabolic crosstalk between cancer and immune cells in cancer niche, the aim is to discover novel strategies of cancer immunotherapy and provide effective guidance for clinical research in cancer treatment. In addition, the review also discusses current challenges such as the inadequacy of relevant diagnostic technologies and the issue of multidrug resistance, and proposes potential solutions including bolstering foundational cancer research, fostering technological innovation, and implementing precision medicine approaches. In-depth research into the metabolic effects of cancer niches can improve cancer treatment outcomes, prolong patients' survival period and enhance their quality of life.
Collapse
Affiliation(s)
- Mingxia Jiang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen 361005, China
| | - Huapan Fang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen 361005, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518000, China
| | - Huayu Tian
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen 361005, China
| |
Collapse
|
8
|
Li W, Li J, Pan C, Lee JS, Kim BS, Gao G. Light-based 3D bioprinting techniques for illuminating the advances of vascular tissue engineering. Mater Today Bio 2024; 29:101286. [PMID: 39435375 PMCID: PMC11492625 DOI: 10.1016/j.mtbio.2024.101286] [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: 06/29/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Vascular tissue engineering faces significant challenges in creating in vitro vascular disease models, implantable vascular grafts, and vascularized tissue/organ constructs due to limitations in manufacturing precision, structural complexity, replicating the composited architecture, and mimicking the mechanical properties of natural vessels. Light-based 3D bioprinting, leveraging the unique advantages of light including high resolution, rapid curing, multi-material adaptability, and tunable photochemistry, offers transformative solutions to these obstacles. With the emergence of diverse light-based 3D bioprinting techniques and innovative strategies, the advances in vascular tissue engineering have been significantly accelerated. This review provides an overview of the human vascular system and its physiological functions, followed by an in-depth discussion of advancements in light-based 3D bioprinting, including light-dominated and light-assisted techniques. We explore the application of these technologies in vascular tissue engineering for creating in vitro vascular disease models recapitulating key pathological features, implantable blood vessel grafts, and tissue analogs with the integration of capillary-like vasculatures. Finally, we provide readers with insights into the future perspectives of light-based 3D bioprinting to revolutionize vascular tissue engineering.
Collapse
Affiliation(s)
- Wei Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology (BIT), Zhuhai 519088, China
| | - Chen Pan
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, China
- School of Mechanical and Equipment Engineering, Hebei University of Engineering, Handan, 050024, China
| | - Jae-Seong Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
9
|
Ramirez DA, Garrott K, Garlitski A, Koop B. Coronary Spasm Due to Pulsed Field Ablation: A State-of-the-Art Review. Pacing Clin Electrophysiol 2024. [PMID: 39494719 DOI: 10.1111/pace.15101] [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: 07/31/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
With the ever-growing population of patients undergoing cardiac ablation with pulsed electric fields, there is a need to understand secondary effects from the therapy. Coronary artery spasm is one such effect that has recently emerged as the subject of further investigation in electrophysiology literature. This review aims to elucidate the basic anatomy underlying vascular spasm due to pulsed electric fields and the effects of irreversible electroporation on coronary arteries. This review also aims to gather the current preclinical and clinical data regarding the physiology and function of coronary arteries following electroporation.
Collapse
Affiliation(s)
- David A Ramirez
- Electrophysiology Research & Development, Boston Scientific Corporation, Marlborough, Massachusetts, USA
| | - Kara Garrott
- Electrophysiology Research & Development, Boston Scientific Corporation, Marlborough, Massachusetts, USA
| | - Ann Garlitski
- Electrophysiology Research & Development, Boston Scientific Corporation, Marlborough, Massachusetts, USA
| | - Brendan Koop
- Electrophysiology Research & Development, Boston Scientific Corporation, Marlborough, Massachusetts, USA
| |
Collapse
|
10
|
Sung JY, Kim SG, Park SY, Kim JR, Choi HC. Telomere stabilization by metformin mitigates the progression of atherosclerosis via the AMPK-dependent p-PGC-1α pathway. Exp Mol Med 2024; 56:1967-1979. [PMID: 39223261 PMCID: PMC11446938 DOI: 10.1038/s12276-024-01297-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: 02/29/2024] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 09/04/2024] Open
Abstract
Telomere dysfunction is a well-known molecular trigger of senescence and has been associated with various age-related diseases, including atherosclerosis. However, the mechanisms involved have not yet been elucidated, and the extent to which telomeres contribute to atherosclerosis is unknown. Therefore, we investigated the mechanism of metformin-induced telomere stabilization and the ability of metformin to inhibit vascular smooth muscle cell (VSMC) senescence caused by advanced atherosclerosis. The present study revealed that metformin inhibited the phenotypes of atherosclerosis and senescence in VSMCs. Metformin increased the phosphorylation of AMPK-dependent PGC-1α and thus increased telomerase activity and the protein level of TERT in OA-treated VSMCs. Mechanistically, the phosphorylation of AMPK and PGC-1α by metformin not only enhanced telomere function but also increased the protein level of TERT, whereas TERT knockdown accelerated the development of atherosclerosis and senescent phenotypes in OA-treated VSMCs regardless of metformin treatment. Furthermore, the in vivo results showed that metformin attenuated the formation of atherosclerotic plaque markers in the aortas of HFD-fed ApoE KO mice. Although metformin did not reduce plaque size, it inhibited the phosphorylation of the AMPK/PGC-1α/TERT signaling cascade, which is associated with the maintenance and progression of plaque formation, in HFD-fed ApoE KO mice. Accordingly, metformin inhibited atherosclerosis-associated phenotypes in vitro and in vivo. These observations show that the enhancement of telomere function by metformin is involved in specific signaling pathways during the progression of atherosclerosis. These findings suggest that telomere stabilization by metformin via the AMPK/p-PGC-1α pathway might provide a strategy for developing therapeutics against vascular diseases such as atherosclerosis.
Collapse
MESH Headings
- Animals
- Male
- Mice
- AMP-Activated Protein Kinases/metabolism
- Atherosclerosis/metabolism
- Atherosclerosis/drug therapy
- Atherosclerosis/pathology
- Atherosclerosis/etiology
- Cellular Senescence/drug effects
- Disease Models, Animal
- Disease Progression
- Metformin/pharmacology
- Metformin/therapeutic use
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
- Phosphorylation/drug effects
- Signal Transduction/drug effects
- Telomerase/metabolism
- Telomerase/genetics
- Telomere/metabolism
- Telomere/drug effects
- Telomere Homeostasis/drug effects
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Jin Young Sung
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu, 42415, Republic of Korea
- Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu, 42415, Republic of Korea
| | - Seul Gi Kim
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu, 42415, Republic of Korea
- Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu, 42415, Republic of Korea
| | - So-Young Park
- Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu, 42415, Republic of Korea
- Department of Physiology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu, 42415, Republic of Korea
| | - Jae-Ryong Kim
- Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu, 42415, Republic of Korea
- Department of Biochemistry and Molecular Biology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu, 42415, Republic of Korea
| | - Hyoung Chul Choi
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu, 42415, Republic of Korea.
- Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu, 42415, Republic of Korea.
| |
Collapse
|
11
|
Dave JM, Chakraborty R, Agyemang A, Ntokou A, Saito J, Ballabh P, Martin KA, Greif DM. Loss of TGFβ-Mediated Repression of Angiopoietin-2 in Pericytes Underlies Germinal Matrix Hemorrhage Pathogenesis. Stroke 2024; 55:2340-2352. [PMID: 39129597 PMCID: PMC11347087 DOI: 10.1161/strokeaha.123.045248] [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: 09/17/2023] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND TGF (transforming growth factor)-β pathway is central to blood-brain barrier development as it regulates cross talk between pericytes and endothelial cells. Murine embryos lacking TGFβ receptor Alk5 (activin receptor-like kinase 5) in brain pericytes (mutants) display endothelial cell hyperproliferation, abnormal vessel morphology, and gross germinal matrix hemorrhage-intraventricular hemorrhage (GMH-IVH), leading to perinatal lethality. Mechanisms underlying how ALK5 signaling in pericytes noncell autonomously regulates endothelial cell behavior remain elusive. METHODS Transcriptomic analysis of human brain pericytes with ALK5 silencing identified differential gene expression. Brain vascular cells isolated from mutant embryonic mice with GMH-IVH and preterm human IVH brain samples were utilized for target validation. Finally, pharmacological and genetic inhibition was used to study the therapeutic effects on GMH-IVH pathology. RESULTS Herein, we establish that the TGFβ/ALK5 pathway robustly represses ANGPT2 (angiopoietin-2) in pericytes via epigenetic remodeling. TGFβ-driven SMAD (suppressor of mothers against decapentaplegic) 3/4 associates with TGIF1 (TGFβ-induced factor homeobox 1) and HDAC (histone deacetylase) 5 to form a corepressor complex at the Angpt2 promoter, resulting in promoter deacetylation and gene repression. Moreover, murine and human germinal matrix vessels display increased ANGPT2 expression during GMH-IVH. Isolation of vascular cells from murine germinal matrix identifies pericytes as a cellular source of excessive ANGPT2. In addition, mutant endothelial cells exhibit higher phosphorylated TIE2 (tyrosine protein kinase receptor). Pharmacological or genetic inhibition of ANGPT2 in mutants improves germinal matrix vessel morphology and attenuates GMH pathogenesis. Importantly, genetic ablation of Angpt2 in mutant pericytes prevents perinatal lethality, prolonging survival. CONCLUSIONS This study demonstrates that TGFβ-mediated ANGPT2 repression in pericytes is critical for maintaining blood-brain barrier integrity and identifies pericyte-derived ANGPT2 as an important pathological target for GMH-IVH.
Collapse
Affiliation(s)
- Jui M. Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Raja Chakraborty
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
| | - Alex Agyemang
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aglaia Ntokou
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Junichi Saito
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Praveen Ballabh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kathleen A. Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
12
|
Daisley H, Acco O, Daisley M, George D, Paul L, Rampersad A, Daisley J. COVID-19 shed light on Virchow's law of thrombosis. Autops Case Rep 2024; 14:e2024512. [PMID: 39372069 PMCID: PMC11452080 DOI: 10.4322/acr.2024.512] [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] [Indexed: 10/08/2024]
Abstract
Virchow's law of thrombosis states that thrombosis in a vessel occurs as a combination of the following: (i) injury to the vessel wall, (ii) stasis of blood flow, and (iii) blood hypercoagulability. Injury to the wall includes infection/inflammation and/or injury to the resident cells of the wall. We postulate that in COVID-19, the SARS-CoV-2 virus directly infects the alveolar type II cell or directly or indirectly infects/injures the pericyte, promoting inflammation and interaction with endothelial cells, thereby causing a cascade of events leading to our observation that thrombosis occurred within the walls of the pulmonary vessels and not in the lumen of the vascular circulation.
Collapse
Affiliation(s)
- Hubert Daisley
- General Hospital, Department of Pathology, San Fernando, Trinidad and Tobago
- Scarborough General Hospital, Department of Pathology, Signal Hill, Trinidad and Tobago
| | - Oneka Acco
- The University of the West Indies, Department of Pathology, Mona, Jamaica
| | - Martina Daisley
- Princes Alexandra Hospital, Accident and Emergency Department The Valley, Anguilla
| | - Dennecia George
- Scarborough General Hospital, Department of Pathology, Signal Hill, Trinidad and Tobago
| | - Lilly Paul
- The University of the West Indies, Department of Pathology, Mona, Jamaica
| | - Arlene Rampersad
- General Hospital, Department of Pathology, San Fernando, Trinidad and Tobago
| | - Johann Daisley
- Scarborough General Hospital, Department of Pathology, Signal Hill, Trinidad and Tobago
| |
Collapse
|
13
|
Choi YE, Yang JM, Jeong CW, Shin S, Park J, Lee K, Cho JH. Prunus yedoensis Bark Downregulates the Expression of Cell Adhesion Molecules in Human Endothelial Cell Lines and Relaxes Blood Vessels in Rat Aortic Rings. Pharmaceuticals (Basel) 2024; 17:926. [PMID: 39065776 PMCID: PMC11279544 DOI: 10.3390/ph17070926] [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: 06/13/2024] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The incidence of cardiovascular diseases, such as high blood pressure, is increasing worldwide, owing to population aging and irregular lifestyle habits. Previous studies have reported the vasorelaxant effects of Prunus yedoensis bark methanol extract. However, various solvent extracts of P. yedoensis bark and their vascular relaxation mechanisms have not been sufficiently studied. We prepared extracts of P. yedoensis bark using various solvents (water, 30% ethanol, and 70% ethanol). P. yedoensis bark 30% ethanol extract (PYB-30E) decreased the expression of vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), and E-selectin in human umbilical vein endothelial cells (HUVECs) activated with 200 ng/mL TNF-α. Additionally, PYB-30E showed vasodilatory effects on isolated rat aortic rings. This was confirmed to be the result of the activation of the NO/cGMP pathway, regulation of non-selective calcium-activated K+ channels, and calcium channel blockade. Additionally, PYB-30E significantly reduced systolic and diastolic blood pressure in spontaneously hypertensive rats (SHR). Taken together, our results indicated that PYB-30E is a candidate functional material with preventive and therapeutic effects against hypertension.
Collapse
Affiliation(s)
- Ye Eun Choi
- Haram Central Research Institute, Cheongju 28160, Republic of Korea; (Y.E.C.); (J.M.Y.); (C.W.J.)
| | - Jung Mo Yang
- Haram Central Research Institute, Cheongju 28160, Republic of Korea; (Y.E.C.); (J.M.Y.); (C.W.J.)
| | - Chae Won Jeong
- Haram Central Research Institute, Cheongju 28160, Republic of Korea; (Y.E.C.); (J.M.Y.); (C.W.J.)
| | - Sujin Shin
- Department of Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Junkyu Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Kyungjin Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Ju Hyun Cho
- Haram Central Research Institute, Cheongju 28160, Republic of Korea; (Y.E.C.); (J.M.Y.); (C.W.J.)
| |
Collapse
|
14
|
Łopianiak I, Kawecka A, Civelek M, Wojasiński M, Cicha I, Ciach T, Butruk-Raszeja BA. Characterization of Blow-Spun Polyurethane Scaffolds-Influence of Fiber Alignment and Fiber Diameter on Pericyte Growth. ACS Biomater Sci Eng 2024; 10:4388-4399. [PMID: 38856968 PMCID: PMC11234331 DOI: 10.1021/acsbiomaterials.4c00051] [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: 06/11/2024]
Abstract
In this study, fibrous polyurethane (PU) materials with average fiber diameter of 200, 500, and 1000 nm were produced using a solution blow spinning (SBS) process. The effects of the rotation speed of the collector (in the range of 200-25 000 rpm) on the fiber alignment and diameter were investigated. The results showed that fiber alignment was influenced by the rotation speed of the collector, and such alignment was possible when the fiber diameter was within a specific range. Homogeneously oriented fibers were obtained only for a fiber diameter ≥500 nm. Moreover, the changes in fiber orientation and fiber diameter (resulting from changes in the rotation speed of the collector) were more noticeable for materials with an average fiber diameter of 1000 nm in comparison to 500 nm, which suggests that the larger the fiber diameter, the better the controlled architectures that can be obtained. The porosity of the produced scaffolds was about 65-70%, except for materials with a fiber diameter of 1000 nm and aligned fibers, which had a higher porosity (76%). Thus, the scaffold pore size increased with increasing fiber diameter but decreased with increasing fiber alignment. The mechanical properties of fibrous materials strongly depend on the direction of stretching, whereby the fiber orientation influences the mechanical strength only for materials with a fiber diameter of 1000 nm. Furthermore, the fiber diameter and alignment affected the pericyte growth. Significant differences in cell growth were observed after 7 days of cell culture between materials with a fiber diameter of 1000 nm (cell coverage 96-99%) and those with a fiber diameter of 500 nm (cell coverage 70-90%). By appropriately setting the SBS process parameters, scaffolds can be easily adapted to the cell requirements, which is of great importance in producing complex 3D structures for guided tissue regeneration.
Collapse
Affiliation(s)
- Iwona Łopianiak
- Laboratory of Biomedical Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, Warsaw 00-645, Poland
- Doctoral School of Warsaw University of Technology, Plac Politechniki 1, Warsaw 00-661, Poland
| | - Aleksandra Kawecka
- Laboratory of Biomedical Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, Warsaw 00-645, Poland
| | - Mehtap Civelek
- Section of Experimental Oncology und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, ENT-Department, Universitätsklinikum, GluckstraBe 10a, Erlangen 91054, Germany
| | - Michał Wojasiński
- Laboratory of Biomedical Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, Warsaw 00-645, Poland
| | - Iwona Cicha
- Section of Experimental Oncology und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, ENT-Department, Universitätsklinikum, GluckstraBe 10a, Erlangen 91054, Germany
| | - Tomasz Ciach
- Laboratory of Biomedical Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, Warsaw 00-645, Poland
| | - Beata A Butruk-Raszeja
- Laboratory of Biomedical Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, Warsaw 00-645, Poland
| |
Collapse
|
15
|
Yrigoin K, Davis GE. Selective mural cell recruitment of pericytes to networks of assembling endothelial cell-lined tubes. Front Cell Dev Biol 2024; 12:1389607. [PMID: 38961866 PMCID: PMC11219904 DOI: 10.3389/fcell.2024.1389607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/30/2024] [Indexed: 07/05/2024] Open
Abstract
Mural cells are critically important for the development, maturation, and maintenance of the blood vasculature. Pericytes are predominantly observed in capillaries and venules, while vascular smooth muscle cells (VSMCs) are found in arterioles, arteries, and veins. In this study, we have investigated functional differences between human pericytes and human coronary artery smooth muscle cells (CASMCs) as a model VSMC type. We compared the ability of these two mural cells to invade three-dimensional (3D) collagen matrices, recruit to developing human endothelial cell (EC)-lined tubes in 3D matrices and induce vascular basement membrane matrix assembly around these tubes. Here, we show that pericytes selectively invade, recruit, and induce basement membrane deposition on EC tubes under defined conditions, while CASMCs fail to respond equivalently. Pericytes dramatically invade 3D collagen matrices in response to the EC-derived factors, platelet-derived growth factor (PDGF)-BB, PDGF-DD, and endothelin-1, while minimal invasion occurs with CASMCs. Furthermore, pericytes recruit to EC tube networks, and induce basement membrane deposition around assembling EC tubes (narrow and elongated tubes) when these cells are co-cultured. In contrast, CASMCs are markedly less able to perform these functions showing minimal recruitment, little to no basement membrane deposition, with wider and shorter tubes. Our new findings suggest that pericytes demonstrate much greater functional ability to invade 3D matrix environments, recruit to EC-lined tubes and induce vascular basement membrane matrix deposition in response to and in conjunction with ECs.
Collapse
Affiliation(s)
| | - George E. Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| |
Collapse
|
16
|
Luo Y, He F, Zhang Y, Li S, Lu R, Wei X, Huang J. Transcription Factor 21: A Transcription Factor That Plays an Important Role in Cardiovascular Disease. Pharmacology 2024; 109:183-193. [PMID: 38493769 DOI: 10.1159/000536585] [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: 12/05/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND According to the World Health Organisation's Health Report 2019, approximately 17.18 million people die from cardiovascular disease each year, accounting for more than 30% of all global deaths. Therefore, the occurrence of cardiovascular disease is still a global concern. The transcription factor 21 (TCF21) plays an important role in cardiovascular diseases. This article reviews the regulation mechanism of TCF21 expression and activity and focuses on its important role in atherosclerosis in order to contribute to the development of diagnosis and treatment of cardiovascular diseases. SUMMARY TCF21 is involved in the phenotypic regulation of vascular smooth muscle cells (VSMCs), promotes the proliferation and migration of VSMCs, and participates in the activation of inflammatory sequences. Increased proliferation and migration of VSMCs can lead to neointimal hyperplasia after vascular injury. Abnormal hyperplasia of neointima and inflammation are one of the main features of atherosclerosis. Therefore, targeting TCF21 may become a potential treatment for relieving atherosclerosis. KEY MESSAGES TCF21 as a member of basic helix-loop-helix transcription factors regulates cell growth and differentiation by modulating gene expression during the development of different organs and plays an important role in cardiovascular development and disease. VSMCs and cells derived from VSMCs constitute the majority of plaques in atherosclerosis. TCF21 plays a key role in regulation of VSMCs' phenotype, thus accelerating atherogenesis in the early stage. However, TCF21 enhances plaque stability in late-stage atherosclerosis. The dual role of TCF21 should be considered in the translational medicine.
Collapse
Affiliation(s)
- Yaqian Luo
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China,
| | - Fangzhou He
- Department of Anaesthesia, Chuanshan College, University of South China, Hengyang, China
| | - Yifang Zhang
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| | - Shufan Li
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Ruirui Lu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Xing Wei
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| | - Ji Huang
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
17
|
Fraile-Martinez O, De Leon-Oliva D, Boaru DL, De Castro-Martinez P, Garcia-Montero C, Barrena-Blázquez S, García-García J, García-Honduvilla N, Alvarez-Mon M, Lopez-Gonzalez L, Diaz-Pedrero R, Guijarro LG, Ortega MA. Connecting epigenetics and inflammation in vascular senescence: state of the art, biomarkers and senotherapeutics. Front Genet 2024; 15:1345459. [PMID: 38469117 PMCID: PMC10925776 DOI: 10.3389/fgene.2024.1345459] [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: 11/27/2023] [Accepted: 02/15/2024] [Indexed: 03/13/2024] Open
Abstract
Vascular diseases pose major health challenges, and understanding their underlying molecular mechanisms is essential to advance therapeutic interventions. Cellular senescence, a hallmark of aging, is a cellular state characterized by cell-cycle arrest, a senescence-associated secretory phenotype macromolecular damage, and metabolic dysregulation. Vascular senescence has been demonstrated to play a key role in different vascular diseases, such as atherosclerosis, peripheral arterial disease, hypertension, stroke, diabetes, chronic venous disease, and venous ulcers. Even though cellular senescence was first described in 1961, significant gaps persist in comprehending the epigenetic mechanisms driving vascular senescence and its subsequent inflammatory response. Through a comprehensive analysis, we aim to elucidate these knowledge gaps by exploring the network of epigenetic alterations that contribute to vascular senescence. In addition, we describe the consequent inflammatory cascades triggered by these epigenetic modifications. Finally, we explore translational applications involving biomarkers of vascular senescence and the emerging field of senotherapy targeting this biological process.
Collapse
Affiliation(s)
- Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Joaquin García-García
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, Alcala deHenares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala deHenares, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala deHenares, Spain
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, Alcala deHenares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, Alcala deHenares, Spain
| |
Collapse
|
18
|
Ibarrola J, Jaffe IZ. The Mineralocorticoid Receptor in the Vasculature: Friend or Foe? Annu Rev Physiol 2024; 86:49-70. [PMID: 37788489 DOI: 10.1146/annurev-physiol-042022-015223] [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: 10/05/2023]
Abstract
Originally described as the renal aldosterone receptor that regulates sodium homeostasis, it is now clear that mineralocorticoid receptors (MRs) are widely expressed, including in vascular endothelial and smooth muscle cells. Ample data demonstrate that endothelial and smooth muscle cell MRs contribute to cardiovascular disease in response to risk factors (aging, obesity, hypertension, atherosclerosis) by inducing vasoconstriction, vascular remodeling, inflammation, and oxidative stress. Extrapolating from its role in disease, evidence supports beneficial roles of vascular MRs in the context of hypotension by promoting inflammation, wound healing, and vasoconstriction to enhance survival from bleeding or sepsis. Advances in understanding how vascular MRs become activated are also reviewed, describing transcriptional, ligand-dependent, and ligand-independent mechanisms. By synthesizing evidence describing how vascular MRs convert cardiovascular risk factors into disease (the vascular MR as a foe), we postulate that the teleological role of the MR is to coordinate responses to hypotension (the MR as a friend).
Collapse
Affiliation(s)
- Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA;
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA;
| |
Collapse
|
19
|
An O, Deppermann C. Platelet lifespan and mechanisms for clearance. Curr Opin Hematol 2024; 31:6-15. [PMID: 37905750 DOI: 10.1097/moh.0000000000000792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
PURPOSE OF REVIEW Activated or aged platelets are removed from circulation under (patho)physiologic conditions, the exact mechanism of platelet clearance under such conditions remains unclear and are currently being investigated. This review focuses on recent findings and controversies regarding platelet clearance and the disruption of platelet life cycle. RECENT FINDINGS The platelet life span is determined by glycosylation of platelet surface receptors with sialic acid. Recently, it was shown that platelet activation and granule release leads to desialylation of glycans and accelerated clearance of platelets under pathological conditions. This phenomenon was demonstrated to be a main reason for thrombocytopenia being a complication in several infections and immune disorders. SUMMARY Although we have recently gained some insight into how aged platelets are cleared from circulation, we are still not seeing the full picture. Further investigations of the platelet clearance pathways under pathophysiologic conditions are needed as well as studies to unravel the connection between platelet clearance and platelet production.
Collapse
Affiliation(s)
- Olga An
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | | |
Collapse
|
20
|
Lin A, Brittan M, Baker AH, Dimmeler S, Fisher EA, Sluimer JC, Misra A. Clonal Expansion in Cardiovascular Pathology. JACC Basic Transl Sci 2024; 9:120-144. [PMID: 38362345 PMCID: PMC10864919 DOI: 10.1016/j.jacbts.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 02/17/2024]
Abstract
Clonal expansion refers to the proliferation and selection of advantageous "clones" that are better suited for survival in a Darwinian manner. In recent years, we have greatly enhanced our understanding of cell clonality in the cardiovascular context. However, our knowledge of the underlying mechanisms behind this clonal selection is still severely limited. There is a transpiring pattern of clonal expansion of smooth muscle cells and endothelial cells-and, in some cases, macrophages-in numerous cardiovascular diseases irrespective of their differing microenvironments. These findings indirectly suggest the possible existence of stem-like vascular cells which are primed to respond during disease. Subsequent clones may undergo further phenotypic changes to adopt either protective or detrimental roles. By investigating these clone-forming vascular cells, we may be able to harness this inherent clonal nature for future therapeutic intervention. This review comprehensively discusses what is currently known about clonal expansion across the cardiovascular field. Comparisons of the clonal nature of vascular cells in atherosclerosis (including clonal hematopoiesis of indeterminate potential), pulmonary hypertension, aneurysm, blood vessel injury, ischemia- and tumor-induced angiogenesis, and cerebral cavernous malformations are evaluated. Finally, we discuss the potential clinical implications of these findings and propose that proper understanding and specific targeting of these clonal cells may provide unique therapeutic options for the treatment of these cardiovascular conditions.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew H. Baker
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- CARIM School for Cardiovascular Sciences, Department of Pathology, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Edward A. Fisher
- Department of Medicine/Division of Cardiology, New York University Grossman School of Medicine, New York, New York, USA
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York, USA
| | - Judith C. Sluimer
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- CARIM School for Cardiovascular Sciences, Department of Pathology, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
21
|
Pereira M, Pinto J, Arteaga B, Guerra A, Jorge RN, Monteiro FJ, Salgado CL. A Comprehensive Look at In Vitro Angiogenesis Image Analysis Software. Int J Mol Sci 2023; 24:17625. [PMID: 38139453 PMCID: PMC10743557 DOI: 10.3390/ijms242417625] [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: 11/16/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
One of the complex challenges faced presently by tissue engineering (TE) is the development of vascularized constructs that accurately mimic the extracellular matrix (ECM) of native tissue in which they are inserted to promote vessel growth and, consequently, wound healing and tissue regeneration. TE technique is characterized by several stages, starting from the choice of cell culture and the more appropriate scaffold material that can adequately support and supply them with the necessary biological cues for microvessel development. The next step is to analyze the attained microvasculature, which is reliant on the available labeling and microscopy techniques to visualize the network, as well as metrics employed to characterize it. These are usually attained with the use of software, which has been cited in several works, although no clear standard procedure has been observed to promote the reproduction of the cell response analysis. The present review analyzes not only the various steps previously described in terms of the current standards for evaluation, but also surveys some of the available metrics and software used to quantify networks, along with the detection of analysis limitations and future improvements that could lead to considerable progress for angiogenesis evaluation and application in TE research.
Collapse
Affiliation(s)
- Mariana Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Jéssica Pinto
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Belén Arteaga
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Faculty of Medicine, University of Granada, Parque Tecnológico de la Salud, Av. de la Investigación 11, 18016 Granada, Spain
| | - Ana Guerra
- INEGI—Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, 4200-465 Porto, Portugal; (A.G.); (R.N.J.)
| | - Renato Natal Jorge
- INEGI—Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, 4200-465 Porto, Portugal; (A.G.); (R.N.J.)
- LAETA—Laboratório Associado de Energia, Transportes e Aeronáutica, Universidade do Porto, 4200-165 Porto, Portugal
- FEUP—Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e de Materiais, Universidade do Porto, 4200-165 Porto, Portugal
| | - Fernando Jorge Monteiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- FEUP—Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e de Materiais, Universidade do Porto, 4200-165 Porto, Portugal
- PCCC—Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
| | - Christiane Laranjo Salgado
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
22
|
Cheng L, Yue H, Zhang H, Liu Q, Du L, Liu X, Xie J, Shen Y. The influence of microenvironment stiffness on endothelial cell fate: Implication for occurrence and progression of atherosclerosis. Life Sci 2023; 334:122233. [PMID: 37918628 DOI: 10.1016/j.lfs.2023.122233] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
Atherosclerosis, the primary cause of cardiovascular diseases (CVDs), is characterized by phenotypic changes in fibrous proliferation, chronic inflammation and lipid accumulation mediated by vascular endothelial cells (ECs) and vascular smooth muscle cells (SMCs) which are correlated with the stiffening and ectopic remodeling of local extracellular matrix (ECM). The native residents, ECs and SMCs, are not only affected by various chemical factors including inflammatory mediators and chemokines, but also by a range of physical stimuli, such as shear stress and ECM stiffness, presented in the microenvironmental niche. Especially, ECs, as a semi-selective barrier, can sense mechanical forces, respond quickly to changes in mechanical loading and provide context-specific adaptive responses to restore homeostasis. However, blood arteries undergo stiffening and lose their elasticity with age. Reports have shown that the ECM stiffening could influence EC fate by changing the cell adhesion, spreading, proliferation, cell to cell contact, migration and even communication with SMCs. The cell behaviour changes mediated by ECM stiffening are dependent on the activation of a signaling cascade of mechanoperception and mechanotransduction. Although the substantial evidence directly indicates the importance of ECM stiffening on the native ECs, the understanding about this complex interplay is still largely limited. In this review, we systematically summarize the roles of ECM stiffening on the behaviours of endothelial cells and elucidate the underlying details in biological mechanism, aiming to provide the process of how ECs integrate ECM mechanics and the highlights for bioaffinity of tissue-specific engineered scaffolds.
Collapse
Affiliation(s)
- Lin Cheng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Hongyan Yue
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Huaiyi Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Qiao Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Lingyu Du
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; JinFeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
23
|
Zhang K, Yue J, Yin L, Chen J, Chen Y, Hu L, Shen J, Yu N, Gong Y, Liu Z. Comprehensive bioinformatics analysis revealed potential key genes and pathways underlying abdominal aortic aneurysm. Comput Struct Biotechnol J 2023; 21:5423-5433. [PMID: 38022704 PMCID: PMC10665597 DOI: 10.1016/j.csbj.2023.10.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a permanent, asymptomatic segmental dilatation of the abdominal aorta, with a high mortality risk upon rupture. Identification of potential key genes and pathways may help to develop curative drugs for AAA. We conducted RNA-seq on abdominal aortic tissues from both AAA patients and normal individuals as a control group. Integrated bioinformatic analysis was subsequently performed to comprehensively reveal potential key genes and pathways. A total of 1148 differential expressed genes (DEGs) (631 up-regulated and 517 down-regulated) were identified in our study. Gene Ontology (GO) analysis revealed enrichment in terms related to extracellular matrix organization, while KEGG analysis indicated enrichment in hematopoietic cell lineage and ECM-receptor interaction. Protein-protein interaction (PPI) network analysis revealed several candidate key genes, and differential expression of 6 key genes (CXCL8, CCL2, PTGS2, SELL, CCR7, and CXCL1) was validated by Gene Expression Omnibus (GEO) datasets. Receiver operating characteristic curve (ROC) analysis demonstrated these genes' high discriminatory ability between AAA and normal tissues. Immunohistochemistry indicated that several key genes were highly expressed in AAA tissues. Single-cell RNA sequencing revealed differential distribution patterns of these identified key genes among various cell types. 26 potential drugs linked to our key genes were found through DGIdb. Overall, our study provides a comprehensive evaluation of potential key genes and pathways in AAA, which could pave the way for the development of curative pharmacological therapies.
Collapse
Affiliation(s)
- Kaijie Zhang
- Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Jianing Yue
- Department of Vascular Surgery, Zhongshan Hospital of Fudan University School of Medicine, Shanghai 200032, China
| | - Li Yin
- Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Jinyi Chen
- Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Yunlu Chen
- Clinical Research Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Lanting Hu
- Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Jian Shen
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Naiji Yu
- Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Yunxia Gong
- Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Zhenjie Liu
- Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| |
Collapse
|
24
|
Palladino S, Schwab A, Copes F, D'Este M, Candiani G, Mantovani D. Development of a hyaluronic acid-collagen bioink for shear-induced fibers and cells alignment. Biomed Mater 2023; 18:065017. [PMID: 37751763 DOI: 10.1088/1748-605x/acfd77] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/26/2023] [Indexed: 09/28/2023]
Abstract
Human tissues are characterized by complex composition and cellular and extracellular matrix (ECM) organization at microscopic level. In most of human tissues, cells and ECM show an anisotropic arrangement, which confers them specific properties.In vitro, the ability to closely mimic this complexity is limited. However, in the last years, extrusion bioprinting showed a certain potential for aligning cells and biomolecules, due to the application of shear stress during the bio-fabrication process. In this work, we propose a strategy to combine collagen (col) with tyramine-modified hyaluronic acid (THA) to obtain a printable col-THA bioink for extrusion bioprinting, solely-based on natural-derived components. Collagen fibers formation within the hybrid hydrogel, as well as collagen distribution and spatial organization before and after printing, were studied. For the validation of the biological outcome, fibroblasts were selected as cellular model and embedded in the col-THA matrix. Cell metabolic activity and cell viability, as well as cell distribution and alignment, were studied in the bioink before and after bioprinting. Results demonstrated successful collagen fibers formation within the bioink, as well as collagen anisotropic alignment along the printing direction. Furthermore, results revealed suitable biological properties, with a slightly reduced metabolic activity at day 1, fully recovered within the first 3 d post-cell embedding. Finally, results showed fibroblasts elongation and alignment along the bioprinting direction. Altogether, results validated the potential to obtain collagen-based bioprinted constructs, with both cellular and ECM anisotropy, without detrimental effects of the fabrication process on the biological outcome. This bioink can be potentially used for a wide range of applications in tissue engineering and regenerative medicine in which anisotropy is required.
Collapse
Affiliation(s)
- Sara Palladino
- Laboratory for Biomaterials and Bioengineering, CRC-Tier I, Dept Min-Met-Materials Eng and Regenerative Medicine, CHU de Québec, Laval University, Quebec City, Canada
- genT_LΛB, Department of Chemistry, Materials and Chemical Engineering 'G. Natta', Politecnico di Milano, Milan, Italy
| | | | - Francesco Copes
- Laboratory for Biomaterials and Bioengineering, CRC-Tier I, Dept Min-Met-Materials Eng and Regenerative Medicine, CHU de Québec, Laval University, Quebec City, Canada
| | | | - Gabriele Candiani
- genT_LΛB, Department of Chemistry, Materials and Chemical Engineering 'G. Natta', Politecnico di Milano, Milan, Italy
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, CRC-Tier I, Dept Min-Met-Materials Eng and Regenerative Medicine, CHU de Québec, Laval University, Quebec City, Canada
| |
Collapse
|
25
|
Yang S, Zeng Z, Yuan Q, Chen Q, Wang Z, Xie H, Liu J. Vascular calcification: from the perspective of crosstalk. MOLECULAR BIOMEDICINE 2023; 4:35. [PMID: 37851172 PMCID: PMC10584806 DOI: 10.1186/s43556-023-00146-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
Vascular calcification (VC) is highly correlated with cardiovascular disease morbidity and mortality, but anti-VC treatment remains an area to be tackled due to the ill-defined molecular mechanisms. Regardless of the type of VC, it does not depend on a single cell but involves multi-cells/organs to form a complex cellular communication network through the vascular microenvironment to participate in the occurrence and development of VC. Therefore, focusing only on the direct effect of pathological factors on vascular smooth muscle cells (VSMCs) tends to overlook the combined effect of other cells and VSMCs, including VSMCs-VSMCs, ECs-VMSCs, Macrophages-VSMCs, etc. Extracellular vesicles (EVs) are a collective term for tiny vesicles with a membrane structure that are actively secreted by cells, and almost all cells secrete EVs. EVs docked on the surface of receptor cells can directly mediate signal transduction or transfer their contents into the cell to elicit a functional response from the receptor cells. They have been proven to participate in the VC process and have also shown attractive therapeutic prospects. Based on the advantages of EVs and the ability to be detected in body fluids, they may become a novel therapeutic agent, drug delivery vehicle, diagnostic and prognostic biomarker, and potential therapeutic target in the future. This review focuses on the new insight into VC molecular mechanisms from the perspective of crosstalk, summarizes how multi-cells/organs interactions communicate via EVs to regulate VC and the emerging potential of EVs as therapeutic methods in VC. We also summarize preclinical experiments on crosstalk-based and the current state of clinical studies on VC-related measures.
Collapse
Affiliation(s)
- Shiqi Yang
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaolin Zeng
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qing Yuan
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qian Chen
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hui Xie
- Department of Orthopaedics, Movement System Injury and Repair Research Centre, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
26
|
Lin PK, Davis GE. Extracellular Matrix Remodeling in Vascular Disease: Defining Its Regulators and Pathological Influence. Arterioscler Thromb Vasc Biol 2023; 43:1599-1616. [PMID: 37409533 PMCID: PMC10527588 DOI: 10.1161/atvbaha.123.318237] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023]
Abstract
Because of structural and cellular differences (ie, degrees of matrix abundance and cross-linking, mural cell density, and adventitia), large and medium-sized vessels, in comparison to capillaries, react in a unique manner to stimuli that induce vascular disease. A stereotypical vascular injury response is ECM (extracellular matrix) remodeling that occurs particularly in larger vessels in response to injurious stimuli, such as elevated angiotensin II, hyperlipidemia, hyperglycemia, genetic deficiencies, inflammatory cell infiltration, or exposure to proinflammatory mediators. Even with substantial and prolonged vascular damage, large- and medium-sized arteries, persist, but become modified by (1) changes in vascular wall cellularity; (2) modifications in the differentiation status of endothelial cells, vascular smooth muscle cells, or adventitial stem cells (each can become activated); (3) infiltration of the vascular wall by various leukocyte types; (4) increased exposure to critical growth factors and proinflammatory mediators; and (5) marked changes in the vascular ECM, that remodels from a homeostatic, prodifferentiation ECM environment to matrices that instead promote tissue reparative responses. This latter ECM presents previously hidden matricryptic sites that bind integrins to signal vascular cells and infiltrating leukocytes (in coordination with other mediators) to proliferate, invade, secrete ECM-degrading proteinases, and deposit injury-induced matrices (predisposing to vessel wall fibrosis). In contrast, in response to similar stimuli, capillaries can undergo regression responses (rarefaction). In summary, we have described the molecular events controlling ECM remodeling in major vascular diseases as well as the differential responses of arteries versus capillaries to key mediators inducing vascular injury.
Collapse
Affiliation(s)
- Prisca K. Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - George E. Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| |
Collapse
|
27
|
Bokhari SMZ, Hamar P. Vascular Endothelial Growth Factor-D (VEGF-D): An Angiogenesis Bypass in Malignant Tumors. Int J Mol Sci 2023; 24:13317. [PMID: 37686121 PMCID: PMC10487419 DOI: 10.3390/ijms241713317] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Vascular endothelial growth factors (VEGFs) are the key regulators of vasculogenesis in normal and oncological development. VEGF-A is the most studied angiogenic factor secreted by malignant tumor cells under hypoxic and inflammatory stress, which made VEGF-A a rational target for anticancer therapy. However, inhibition of VEGF-A by monoclonal antibody drugs led to the upregulation of VEGF-D. VEGF-D was primarily described as a lymphangiogenic factor; however, VEGF-D's blood angiogenic potential comparable to VEGF-A has already been demonstrated in glioblastoma and colorectal carcinoma. These findings suggested a role for VEGF-D in facilitating malignant tumor growth by bypassing the anti-VEGF-A antiangiogenic therapy. Owing to its high mitogenic ability, higher affinity for VEGFR-2, and higher expression in cancer, VEGF-D might even be a stronger angiogenic driver and, hence, a better therapeutic target than VEGF-A. In this review, we summarized the angiogenic role of VEGF-D in blood vasculogenesis and its targetability as an antiangiogenic therapy in cancer.
Collapse
Affiliation(s)
| | - Peter Hamar
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary;
| |
Collapse
|
28
|
Ranjbar J, Njoroge W, Gibbins JM, Roach P, Yang Y, Harper AGS. Developing Biomimetic Hydrogels of the Arterial Wall as a Prothrombotic Substrate for In Vitro Human Thrombosis Models. Gels 2023; 9:477. [PMID: 37367147 DOI: 10.3390/gels9060477] [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: 05/10/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Current in vitro thrombosis models utilise simplistic 2D surfaces coated with purified components of the subendothelial matrix. The lack of a realistic humanised model has led to greater study of thrombus formation in in vivo tests in animals. Here we aimed to develop 3D hydrogel-based replicas of the medial and adventitial layers of the human artery to produce a surface that can optimally support thrombus formation under physiological flow conditions. These tissue-engineered medial- (TEML) and adventitial-layer (TEAL) hydrogels were developed by culturing human coronary artery smooth muscle cells and human aortic adventitial fibroblasts within collagen hydrogels, both individually and in co-culture. Platelet aggregation upon these hydrogels was studied using a custom-made parallel flow chamber. When cultured in the presence of ascorbic acid, the medial-layer hydrogels were able to produce sufficient neo-collagen to support effective platelet aggregation under arterial flow conditions. Both TEML and TEAL hydrogels possessed measurable tissue factor activity and could trigger coagulation of platelet-poor plasma in a factor VII-dependent manner. Biomimetic hydrogel replicas of the subendothelial layers of the human artery are effective substrates for a humanised in vitro thrombosis model that could reduce animal experimentation by replacing current in vivo models.
Collapse
Affiliation(s)
- Jacob Ranjbar
- School of Medicine, Keele University, Keele ST5 5BG, UK
| | - Wanjiku Njoroge
- School of Pharmacy & Bioengineering, Keele University, Keele ST5 5BG, UK
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading RG6 6UB, UK
| | - Paul Roach
- Department of Chemistry, School of Science, Loughborough University, Loughborough LE11 3TU, UK
| | - Ying Yang
- School of Pharmacy & Bioengineering, Keele University, Keele ST5 5BG, UK
| | | |
Collapse
|
29
|
Li Y, Tao L, Xu Y, Guo R. Taxifolin ameliorates abdominal aortic aneurysm by preventing inflammation and apoptosis and extracellular matrix degradation via inactivating TLR4/NF-κB axis. Int Immunopharmacol 2023; 119:110197. [PMID: 37098322 DOI: 10.1016/j.intimp.2023.110197] [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: 09/28/2022] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 04/27/2023]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a serious aortic disease with high mortality. Vascular smooth muscle cells (VSMCs) loss is a prominent feature of AAA. Taxifolin (TXL) is a natural antioxidant polyphenol and possesses therapeutic functions in numerous human diseases. This study aimed to investigate TXL's impact on VSMC phenotype in AAA. METHODS In vitro and in vivo of VSMC injury model was induced by angiotensin II (Ang II). The potential function of TXL on AAA was determined using Cell Counting Kit-8, flow cytometry, Western blot, quantitative reverse transcription-PCR, and enzyme-linked immunosorbent assay. Meanwhile, TXL mechanism on AAA was checked by a series of molecular experiments. Also, TXL function on AAA in vivo was further evaluated using hematoxylin-eosin staining, TUNEL assay, Picric acid-Sirius red staining and immunofluorescence assay in C57BL/6 mice. RESULTS TXL alleviated Ang II-induced VSMC injury mainly by enhancing VSMC proliferation and weakening cell apoptosis, alleviating VSMC inflammation, and reducing extracellular matrix (ECM) degradation of VSMCs. Furthermore, mechanistic studies corroborated that TXL reversed the high levels of Toll-like receptor 4 (TLR4) and p-p65/p65 induced by Ang II. Also, TXL facilitated VSMC proliferation and reduced cell apoptosis, repressed inflammation, and ECM degradation of VSMCs, while these effects were reversed by TLR4 overexpression. In vivo studies further confirmed that TXL owned the function of alleviating AAA, such as alleviating collagen fiber hyperplasia and inflammatory cell infiltration in AAA mice, and repressing inflammation and ECM degradation. CONCLUSION TXL protected VSMCs against Ang II-induced injury through activating TLR4/noncanonical nuclear factor-kappaB(NF-κB).
Collapse
Affiliation(s)
- Yuanmin Li
- Department of Cardio-Thoracic Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China
| | - Lingyun Tao
- Shanghai Laboratory Animal Research Center, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China
| | - Rong Guo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China.
| |
Collapse
|
30
|
Gusev E, Sarapultsev A. Atherosclerosis and Inflammation: Insights from the Theory of General Pathological Processes. Int J Mol Sci 2023; 24:ijms24097910. [PMID: 37175617 PMCID: PMC10178362 DOI: 10.3390/ijms24097910] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Recent advances have greatly improved our understanding of the molecular mechanisms behind atherosclerosis pathogenesis. However, there is still a need to systematize this data from a general pathology perspective, particularly with regard to atherogenesis patterns in the context of both canonical and non-classical inflammation types. In this review, we analyze various typical phenomena and outcomes of cellular pro-inflammatory stress in atherosclerosis, as well as the role of endothelial dysfunction in local and systemic manifestations of low-grade inflammation. We also present the features of immune mechanisms in the development of productive inflammation in stable and unstable plaques, along with their similarities and differences compared to canonical inflammation. There are numerous factors that act as inducers of the inflammatory process in atherosclerosis, including vascular endothelium aging, metabolic dysfunctions, autoimmune, and in some cases, infectious damage factors. Life-critical complications of atherosclerosis, such as cardiogenic shock and severe strokes, are associated with the development of acute systemic hyperinflammation. Additionally, critical atherosclerotic ischemia of the lower extremities induces paracoagulation and the development of chronic systemic inflammation. Conversely, sepsis, other critical conditions, and severe systemic chronic diseases contribute to atherogenesis. In summary, atherosclerosis can be characterized as an independent form of inflammation, sharing similarities but also having fundamental differences from low-grade inflammation and various variants of canonical inflammation (classic vasculitis).
Collapse
Affiliation(s)
- Evgenii Gusev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080 Chelyabinsk, Russia
| |
Collapse
|
31
|
Deng H, Li B, Shen Q, Zhang C, Kuang L, Chen R, Wang S, Ma Z, Li G. Mechanisms of diabetic foot ulceration: A review. J Diabetes 2023; 15:299-312. [PMID: 36891783 PMCID: PMC10101842 DOI: 10.1111/1753-0407.13372] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/23/2023] [Accepted: 02/16/2023] [Indexed: 03/10/2023] Open
Abstract
Diabetic foot ulcers (DFUs) are associated with complex pathogenic factors and are considered a serious complication of diabetes. The potential mechanisms underlying DFUs have been increasingly investigated. Previous studies have focused on the three aspects of diabetic peripheral vascular disease, neuropathy, and wound infections. With advances in technology, researchers have been gradually conducting studies using immune cells, endothelial cells, keratinocytes, and fibroblasts, as they are involved in wound healing. It has been reported that the upregulation or downregulation of molecular signaling pathways is essential for the healing of DFUs. With a recent increase in the awareness of epigenetics, its regulatory role in wound healing has become a much sought-after trend in the treatment of DFUs. This review focuses on four aspects involved in the pathogenesis of DFUs: physiological and pathological mechanisms, cellular mechanisms, molecular signaling pathway mechanisms, and epigenetics. Given the challenge in the treatment of DFUs, we are hopeful that our review will provide new ideas for peers.
Collapse
Affiliation(s)
- Haibo Deng
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Binghui Li
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Qian Shen
- School of Foreign StudiesZhongnan University of Economics and LawWuhanHubeiChina
| | - Chenchen Zhang
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Liwen Kuang
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Ran Chen
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - SiYuan Wang
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - ZhiQiang Ma
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Gongchi Li
- Department of Hand Surgery, Union Hospital affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
32
|
Siddhartha R, Garg M. Interplay Between Extracellular Matrix Remodeling and Angiogenesis in Tumor Ecosystem. Mol Cancer Ther 2023; 22:291-305. [PMID: 36861362 DOI: 10.1158/1535-7163.mct-22-0595] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/27/2022] [Accepted: 12/16/2022] [Indexed: 03/03/2023]
Abstract
Studying the complex mechanisms of tumorigenesis and examining the interactions of neoplastic cells within tumor ecosystem are critical to explore the possibility of effective cancer treatment modalities. Dynamic tumor ecosystem is constantly evolving and is composed of tumor cells, extracellular matrix (ECM), secreted factors, and stromal cancer-associated fibroblasts (CAF), pericytes, endothelial cells (EC), adipocytes, and immune cells. ECM remodeling by synthesis, contraction, and/or proteolytic degradation of ECM components and release of matrix-sequestered growth factors create a microenvironment that promotes EC proliferation, migration, and angiogenesis. Stromal CAFs release multiple angiogenic cues (angiogenic growth factors, cytokines, and proteolytic enzymes) which interact with ECM proteins, thus contribute to enhance proangiogenic/promigratory properties and support aggressive tumor growth. Targeting angiogenesis brings about vascular changes including reduced adherence junction proteins, basement membrane and pericyte coverage, and increased leakiness. This facilitates ECM remodeling, metastatic colonization and chemoresistance. Owing to significant role of denser and stiffer ECM in inducing chemoresistance, direct or indirect targeting of ECM components is being reported as major axis of anticancer treatment. Exploring the agents targeting angiogenesis and ECM in a context specific manner may lead to reduced tumor burden by promoting conventional therapeutic effectiveness and overcoming the hurdles of therapy resistance.
Collapse
Affiliation(s)
- Rohit Siddhartha
- Department of Biochemistry, University of Lucknow, Lucknow, India
| | - Minal Garg
- Department of Biochemistry, University of Lucknow, Lucknow, India
| |
Collapse
|
33
|
Zhou J, Gummi MR, Greco A, Babic M, Herrmann J, Kandil FI, van der Giet M, Tölle M, Schuchardt M. Biomechanical Properties of the Aortic Wall: Changes during Vascular Calcification. Biomedicines 2023; 11:211. [PMID: 36672718 PMCID: PMC9855732 DOI: 10.3390/biomedicines11010211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/17/2023] Open
Abstract
Medial vascular calcification (MAC) is characterized by the deposition of hydroxyapatite (HAP) in the medial layer of the vessel wall, leading to disruption of vessel integrity and vascular stiffness. Because currently no direct therapeutic interventions for MAC are available, studying the MAC pathogenesis is of high research interest. Several methods exist to measure and describe the pathophysiological processes in the vessel wall, such as histological staining and gene expression. However, no method describing the physiological properties of the arterial wall is currently available. This study aims to close that gap and validate a method to measure the biomechanical properties of the arterial wall during vascular calcification. Therefore, a stress-stretch curve is monitored using small-vessel-myography upon ex vivo calcification of rat aortic tissue. The measurement of biomechanical properties could help to gain further insights into vessel integrity during calcification progression.
Collapse
Affiliation(s)
- Jinwen Zhou
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Manasa Reddy Gummi
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Anna Greco
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Milen Babic
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Jaqueline Herrmann
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Farid I. Kandil
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Social Medicine, Epidemiology and Health Economics, Luisenstraße 57, 10117 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Pediatric Oncology/Hematology, Otto-Heubner Centre for Pediatric and Adolescent Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Markus van der Giet
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Markus Tölle
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Mirjam Schuchardt
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
34
|
Yin Y, Huang C, Wang Z, Huang P, Qin S. Identification of cellular heterogeneity and key signaling pathways associated with vascular remodeling and calcification in young and old primate aortas based on single-cell analysis. Aging (Albany NY) 2022; 15:982-1003. [PMID: 36566020 PMCID: PMC10008505 DOI: 10.18632/aging.204442] [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: 09/30/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Aging of the vascular system is the main cause of many cardiovascular diseases. The structure and function of the blood vessel wall change with aging. To prevent age-related cardiovascular diseases, it is essential to understand the cellular heterogeneity of vascular wall and changes of cellular communication among cell subpopulations during aging. Here, using published single-cell RNA sequencing datasets of young and old monkey aortas, we analyzed the heterogeneity of vascular endothelial cells and smooth muscle cells in detail and identified a distinct endothelial cell subpopulation that involved in vascular remodeling and calcification. Moreover, cellular communication that changed with aging was analyzed and we identified a number of signaling pathways that associated with vascular aging. We found that EGF signaling pathway play an essential role in vascular remodeling and calcification of aged aortas. This work provided a better understanding of vascular aging and laid the foundation for prevention of age-related vascular pathologies.
Collapse
Affiliation(s)
- Yehu Yin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China.,Institute of Medicine, Jishou University, Jishou 416000, P.R. China
| | - Congcong Huang
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China
| | - Zidi Wang
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China
| | - Pan Huang
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China.,Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China
| |
Collapse
|
35
|
Ye C, Zheng F, Wu N, Zhu GQ, Li XZ. Extracellular vesicles in vascular remodeling. Acta Pharmacol Sin 2022; 43:2191-2201. [PMID: 35022541 PMCID: PMC9433397 DOI: 10.1038/s41401-021-00846-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022]
Abstract
Vascular remodeling contributes to the development of a variety of vascular diseases including hypertension and atherosclerosis. Phenotypic transformation of vascular cells, oxidative stress, inflammation and vascular calcification are closely associated with vascular remodeling. Extracellular vesicles (EVs) are naturally released from almost all types of cells and can be detected in nearly all body fluids including blood and urine. EVs affect vascular oxidative stress, inflammation, calcification, and lipid plaque formation; and thereby impact vascular remodeling in a variety of cardiovascular diseases. EVs may be used as biomarkers for diagnosis and prognosis, and therapeutic strategies for vascular remodeling and cardiovascular diseases. This review includes a comprehensive analysis of the roles of EVs in the vascular remodeling in vascular diseases, and the prospects of EVs in the diagnosis and treatment of vascular diseases.
Collapse
Affiliation(s)
- Chao Ye
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Fen Zheng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Nan Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China.
| | - Xiu-Zhen Li
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
36
|
Liu Z, Andraska E, Akinbode D, Mars W, Alvidrez RIM. LRP1 in the Vascular Wall. CURRENT PATHOBIOLOGY REPORTS 2022. [DOI: 10.1007/s40139-022-00231-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Wang S, Hu S. The Role of Sirtuins in Osteogenic Differentiation of Vascular Smooth Muscle Cells and Vascular Calcification. Front Cardiovasc Med 2022; 9:894692. [PMID: 35722093 PMCID: PMC9198215 DOI: 10.3389/fcvm.2022.894692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular calcification (VC) is a common pathological change in many chronic diseases, such as diabetes and chronic kidney disease. It is mainly deposited in the intima and media of vessels in the form of hydroxyapatite. Recently, a lot of research has been performed to show that VC is associated with various cellular stresses, such as hyperphosphate, hyperglycemia and oxidative stress. Unfortunately, our understanding of the pathogenesis of calcification is far from comprehensive. Sirtuins belong to a family of class III highly conserved deacetylases that are involved in the regulation of biological and cellular processes including mitochondrial biogenesis, metabolism, oxidative stress, inflammatory response, DNA repair, etc. Numerous studies have shown that sirtuins might play protective roles in VC, and restoring the activity of sirtuins may be a potentially effective treatment for VC. However, the exact mechanism of their vascular protection remains unclear. Here, we reviewed the roles of sirtuins in the osteogenic transformation of vascular smooth muscle cells and the development of VC. We also elucidated the applications of sirtuins agonists for the treatment of VC.
Collapse
Affiliation(s)
- Shuangshuang Wang
- Department of Cardiology, The First People's Hospital of Wenling (The Affiliated Wenling Hospital of Wenzhou Medical University), Wenling, China
| | - Siwang Hu
- The Orthopedic Center, The First People's Hospital of Wenling (The Affiliated Wenling Hospital of Wenzhou Medical University), Wenling, China
- *Correspondence: Siwang Hu
| |
Collapse
|
38
|
Hassan MAA, Sayed RKA, Abdelsabour-Khalaf M, Abd-Elhafez EA, Anel-Lopez L, Riesco MF, Ortega-Ferrusola C, Montes-Garrido R, Neila-Montero M, Anel L, Alvarez M. Morphological and ultrasonographic characterization of the three zones of supratesticular region of testicular artery in Assaf rams. Sci Rep 2022; 12:8334. [PMID: 35585142 PMCID: PMC9117313 DOI: 10.1038/s41598-022-12243-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/09/2022] [Indexed: 11/25/2022] Open
Abstract
To fully understand the histological, morphometrical and heamodynamic variations of different supratesticular artery regions, 20 mature and healthy Assaf rams were examined through ultrasound and morphological studies. The testicular artery images of the spermatic cord as shown by B-mode analysis indicated a tortuous pattern along its course toward the testis, although it tends to be less tortuous close to the inguinal ring. Doppler velocimetric values showed a progressive decline in flow velocity, in addition to pulsatility and vessel resistivity when entering the testis, where there were significant differences in the Doppler indices and velocities among the different regions. The peak systolic velocity, pulsatility index and resistive index were higher in the proximal supratesticular artery region, followed by middle and distal ones, while the end diastolic velocity was higher in the distal supratesticular region. The total arterial blood flow and total arterial blood flow rate reported a progressive and significant increase along the testicular cord until entering the testis. Histological examination revealed presence of vasa vasorum in the tunica adventitia, with their diameter is higher in the proximal supratesticular zone than middle and distal ones. Morphometrically, the thickness of the supratesticular artery wall showed a significant decline downward toward the testis; meanwhile, the outer arterial diameter and inner luminal diameter displayed a significant increase distally. The expression of alpha smooth muscle actin and vimentin was higher in the tunica media of the proximal supratesticular artery zone than in middle and distal ones.
Collapse
Affiliation(s)
- Mohamed A A Hassan
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag, 82524, Egypt
| | - Ramy K A Sayed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag, 82524, Egypt
| | - Mohammed Abdelsabour-Khalaf
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | - Enas A Abd-Elhafez
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - L Anel-Lopez
- ITRA-ULE, INDEGSAL, University of León, 24071, León, Spain. .,Department of Veterinary Medicine, Surgery and Anatomy, University of León, 24071, León, Spain.
| | - M F Riesco
- ITRA-ULE, INDEGSAL, University of León, 24071, León, Spain.,Cellular Biology, Department of Molecular Biology, University of León, 24071, León, Spain
| | - C Ortega-Ferrusola
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - R Montes-Garrido
- ITRA-ULE, INDEGSAL, University of León, 24071, León, Spain.,Department of Veterinary Medicine, Surgery and Anatomy, University of León, 24071, León, Spain
| | - M Neila-Montero
- ITRA-ULE, INDEGSAL, University of León, 24071, León, Spain.,Department of Veterinary Medicine, Surgery and Anatomy, University of León, 24071, León, Spain
| | - L Anel
- ITRA-ULE, INDEGSAL, University of León, 24071, León, Spain.,Department of Veterinary Medicine, Surgery and Anatomy, University of León, 24071, León, Spain
| | - M Alvarez
- ITRA-ULE, INDEGSAL, University of León, 24071, León, Spain.,Department of Veterinary Medicine, Surgery and Anatomy, University of León, 24071, León, Spain
| |
Collapse
|
39
|
Misra A, Rehan R, Lin A, Patel S, Fisher EA. Emerging Concepts of Vascular Cell Clonal Expansion in Atherosclerosis. Arterioscler Thromb Vasc Biol 2022; 42:e74-e84. [PMID: 35109671 PMCID: PMC8988894 DOI: 10.1161/atvbaha.121.316093] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clonal expansion is a process that can drive pathogenesis in human diseases, with atherosclerosis being a prominent example. Despite advances in understanding the etiology of atherosclerosis, clonality studies of vascular cells remain in an early stage. Recently, several paradigm-shifting preclinical studies have identified clonal expansion of progenitor cells in the vasculature in response to atherosclerosis. This review provides an overview of cell clonality in atherosclerotic progression, focusing particularly on smooth muscle cells and macrophages. We discuss key findings from the latest research that give insight into the mechanisms by which clonal expansion of vascular cells contributes to disease pathology. The further probing of these mechanisms will provide innovative directions for future progress in the understanding and therapy of atherosclerosis and its associated cardiovascular diseases.
Collapse
Affiliation(s)
- Ashish Misra
- Heart Research Institute, Sydney, NSW 2042, Australia,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Rajan Rehan
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia,Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Alexander Lin
- Heart Research Institute, Sydney, NSW 2042, Australia,School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sanjay Patel
- Heart Research Institute, Sydney, NSW 2042, Australia,Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Edward A Fisher
- Department of Medicine/Division of Cardiology, New York University Grossman School of Medicine, New York, NY, USA,Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
40
|
Ayoub NM, Jaradat SK, Al-Shami KM, Alkhalifa AE. Targeting Angiogenesis in Breast Cancer: Current Evidence and Future Perspectives of Novel Anti-Angiogenic Approaches. Front Pharmacol 2022; 13:838133. [PMID: 35281942 PMCID: PMC8913593 DOI: 10.3389/fphar.2022.838133] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a vital process for the growth and dissemination of solid cancers. Numerous molecular pathways are known to drive angiogenic switch in cancer cells promoting the growth of new blood vessels and increased incidence of distant metastasis. Several angiogenesis inhibitors are clinically available for the treatment of different types of advanced solid cancers. These inhibitors mostly belong to monoclonal antibodies or small-molecule tyrosine kinase inhibitors targeting the classical vascular endothelial growth factor (VEGF) and its receptors. Nevertheless, breast cancer is one example of solid tumors that had constantly failed to respond to angiogenesis inhibitors in terms of improved survival outcomes of patients. Accordingly, it is of paramount importance to assess the molecular mechanisms driving angiogenic signaling in breast cancer to explore suitable drug targets that can be further investigated in preclinical and clinical settings. This review summarizes the current evidence for the effect of clinically available anti-angiogenic drugs in breast cancer treatment. Further, major mechanisms associated with intrinsic or acquired resistance to anti-VEGF therapy are discussed. The review also describes evidence from preclinical and clinical studies on targeting novel non-VEGF angiogenic pathways in breast cancer and several approaches to the normalization of tumor vasculature by targeting pericytes, utilization of microRNAs and extracellular tumor-associate vesicles, using immunotherapeutic drugs, and nanotechnology.
Collapse
Affiliation(s)
- Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
- *Correspondence: Nehad M. Ayoub,
| | - Sara K. Jaradat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Kamal M. Al-Shami
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Amer E. Alkhalifa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| |
Collapse
|
41
|
Shang F, Guo X, Chen Y, Wang C, Gao J, Wen E, Lai B, Bai L. Endothelial MicroRNA-483-3p Is Hypertension-Protective. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3698219. [PMID: 35222797 PMCID: PMC8872655 DOI: 10.1155/2022/3698219] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/26/2022] [Indexed: 12/21/2022]
Abstract
Hypertension is a high-risk factor for developing coronary heart disease and stroke. Endothelial dysfunction and arterial remodeling can lead to increased vascular wall thickness and arterial stiffness. Previous studies showed that microRNA-483 (miR-483) enhances endothelial cell (EC) function. Here, we investigated the protective role of miR-483 in hypertension. Data collected from two patient cohorts showed that the serum miR-483-3p level was associated with the progression of hypertension and positively correlated with vascular function. In cultured ECs, miR-483 targets a number of endothelial dysfunction-related genes, such as transforming growth factor-β (TGF-β), connective tissue growth factor (CTGF), angiotensin-converting enzyme 1 (ACE1), and endothelin-1 (ET-1). Overexpression of miR-483-3p in ECs inhibited Ang II-induced endothelial dysfunction, revealed by the decreased expression of TGF-β, CTGF, ACE1, and ET-1. Furthermore, miR-483-3p secreted from ECs was taken up by smooth muscle cells (SMCs) via the exosome pathway, which also decreased these genes in SMCs. Additionally, telmisartan could increase the aortic and serum levels of miR-483-3p in hypertension patients and spontaneous hypertension rats (SHR). These findings suggest that miR-483-3p exerts a protective effect on EC function during the onset of hypertension and thus may be considered a potential therapeutic target for hypertension-related cardiovascular diseases.
Collapse
Affiliation(s)
- Fenqing Shang
- Translational Medicine Center, Xi'an Chest Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xuan Guo
- Department of Cardiology, Xi'an No. 1 Hospital, Xi'an, Shaanxi, China
- Department of Cardiology, Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi 710061, China
| | - Yueer Chen
- Translational Medicine Center, Xi'an Chest Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Chen Wang
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jie Gao
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ergang Wen
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Baochang Lai
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Liang Bai
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
42
|
Abdominal Aortic Aneurysm Formation with a Focus on Vascular Smooth Muscle Cells. Life (Basel) 2022; 12:life12020191. [PMID: 35207478 PMCID: PMC8880357 DOI: 10.3390/life12020191] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/29/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a lethal degenerative vascular disease that affects, mostly, the elder population, with a high mortality rate (>80%) upon rupture. It features a dilation of the aortic diameter to larger than 30 mm or more than 50%. Diverse pathological processes are involved in the development of AAA, including aortic wall inflammation, elastin breakdown, oxidative stress, smooth muscle cell (SMC) phenotypic switching and dysfunction, and extracellular matrix degradation. With open surgery being the only therapeutic option up to date, the lack of pharmaceutical treatment approach calls for identifying novel and effective targets and further understanding the pathological process of AAA. Both lifestyle and genetic predisposition have an important role in increasing the risk of AAA. Several cell types are closely related to the pathogenesis of AAA. Among them, vascular SMCs (VSMCs) are gaining much attention as a critical contributor for AAA initiation and/or progression. In this review, we summarize what is known about AAA, including the risk factors, the pathophysiology, and the established animal models of AAA. In particular, we focus on the VSMC phenotypic switching and dysfunction in AAA formation. Further understanding the regulation of VSMC phenotypic changes may provide novel therapeutic targets for the treatment or prevention of AAA.
Collapse
|
43
|
Mikołajczyk K, Spyt D, Zielińska W, Żuryń A, Faisal I, Qamar M, Świniarski P, Grzanka A, Gagat M. The Important Role of Endothelium and Extracellular Vesicles in the Cellular Mechanism of Aortic Aneurysm Formation. Int J Mol Sci 2021; 22:ijms222313157. [PMID: 34884962 PMCID: PMC8658239 DOI: 10.3390/ijms222313157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Homeostasis is a fundamental property of biological systems consisting of the ability to maintain a dynamic balance of the environment of biochemical processes. The action of endogenous and exogenous factors can lead to internal balance disorder, which results in the activation of the immune system and the development of inflammatory response. Inflammation determines the disturbances in the structure of the vessel wall, connected with the change in their diameter. These disorders consist of accumulation in the space between the endothelium and the muscle cells of low-density lipoproteins (LDL), resulting in the formation of fatty streaks narrowing the lumen and restricting the blood flow in the area behind the structure. The effect of inflammation may also be pathological dilatation of the vessel wall associated with the development of aneurysms. Described disease entities strongly correlate with the increased migration of immune cells. Recent scientific research indicates the secretion of specific vesicular structures during migration activated by the inflammation. The review focuses on the link between endothelial dysfunction and the inflammatory response and the impact of these processes on the development of disease entities potentially related to the secretion of extracellular vesicles (EVs).
Collapse
Affiliation(s)
- Klaudia Mikołajczyk
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Dominika Spyt
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Wioletta Zielińska
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Agnieszka Żuryń
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Inaz Faisal
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Murtaz Qamar
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Piotr Świniarski
- Department of Urology and Andrology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland;
| | - Alina Grzanka
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
| | - Maciej Gagat
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.M.); (D.S.); (W.Z.); (A.Ż.); (I.F.); (M.Q.); (A.G.)
- Correspondence:
| |
Collapse
|
44
|
S SN, Saha S, Bhattacharjee A. A 2D FSI mathematical model of blood flow to analyze the hyper-viscous effects in atherosclerotic COVID patients. RESULTS IN ENGINEERING 2021; 12:100275. [PMID: 35317220 PMCID: PMC8382661 DOI: 10.1016/j.rineng.2021.100275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/21/2021] [Accepted: 08/22/2021] [Indexed: 06/14/2023]
Abstract
Recent researches on COVID 19 has been extended to analyze the various morphological and anatomical changes in a patient's body due to the invasion of the virus. These latest studies have concluded that there happens a high rise in the viscosity of the blood in a COVID 19 patient, supported by the extensive analysis of the clinical data. In the present paper, a mathematical model in the form of a differential equation system has been proposed to disclose the various changes that occur in the flow across the stenosis of an arterial segment. The consequences of the hyperviscosity of blood on the blood flow characteristics in a stenosed artery are analyzed by solving the model using a finite element method (FEM) solver. A laminar flow coupled with solid mechanics through the Fluid-Structure Interaction (FSI) interface has been studied using an Arbitrary Lagrangian-Eulerian (ALE) method. For the first time, the mathematical model was used to analyze the hyper-viscous flow condition in COVID 19 patients. The present research is mainly based on the numerous clinical reports enlisting the various morphological, hematological, and rheological changes in the blood.
Collapse
Affiliation(s)
- Shankar Narayan S
- CMR Institute of Technology, Bengaluru, 560037, India
- Dayananda Sagar University, Bengaluru, 560068, India
| | - Sunanda Saha
- Vellore Institute of Technology, Vellore, 632014, India
| | | |
Collapse
|
45
|
Omar R, Malfait F, Van Agtmael T. Four decades in the making: Collagen III and mechanisms of vascular Ehlers Danlos Syndrome. Matrix Biol Plus 2021; 12:100090. [PMID: 34849481 PMCID: PMC8609142 DOI: 10.1016/j.mbplus.2021.100090] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/10/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Vascular Ehlers Danlos (vEDS) syndrome is a severe multi-systemic connective tissue disorder characterized by risk of dissection and rupture of the arteries, gastro-intestinal tract and gravid uterus. vEDS is caused by mutations in COL3A1, that encodes the alpha 1 chain of type III collagen, which is a major extracellular matrix component of the vasculature and hollow organs. The first causal mutations were identified in the 1980s but progress in our understanding of the pathomolecular mechanisms has been limited. Recently, the application of more refined animal models combined with global omics approaches has yielded important new insights both in terms of disease mechanisms and potential for therapeutic intervention. However, it is also becoming apparent that vEDS is a complex disorder in terms of its molecular disease mechanisms with a poorly understood allelic and mechanistic heterogeneity. In this brief review we will focus our attention on the disease mechanisms of COL3A1 mutations and vEDS, and recent progress in therapeutic approaches using animal models.
Collapse
Affiliation(s)
- Ramla Omar
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, G12 8QQ, UK
| | - Fransiska Malfait
- Centre for Medical Genetics, Ghent University Hospital, Belgium
- Department of Biomolecular Medicine, Ghent University, Belgium
| | - Tom Van Agtmael
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, G12 8QQ, UK
| |
Collapse
|
46
|
Cellular Crosstalk between Endothelial and Smooth Muscle Cells in Vascular Wall Remodeling. Int J Mol Sci 2021; 22:ijms22147284. [PMID: 34298897 PMCID: PMC8306829 DOI: 10.3390/ijms22147284] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/25/2021] [Accepted: 07/01/2021] [Indexed: 12/24/2022] Open
Abstract
Pathological vascular wall remodeling refers to the structural and functional changes of the vessel wall that occur in response to injury that eventually leads to cardiovascular disease (CVD). Vessel wall are composed of two major primary cells types, endothelial cells (EC) and vascular smooth muscle cells (VSMCs). The physiological communications between these two cell types (EC–VSMCs) are crucial in the development of the vasculature and in the homeostasis of mature vessels. Moreover, aberrant EC–VSMCs communication has been associated to the promotor of various disease states including vascular wall remodeling. Paracrine regulations by bioactive molecules, communication via direct contact (junctions) or information transfer via extracellular vesicles or extracellular matrix are main crosstalk mechanisms. Identification of the nature of this EC–VSMCs crosstalk may offer strategies to develop new insights for prevention and treatment of disease that curse with vascular remodeling. Here, we will review the molecular mechanisms underlying the interplay between EC and VSMCs. Additionally, we highlight the potential applicable methodologies of the co-culture systems to identify cellular and molecular mechanisms involved in pathological vascular wall remodeling, opening questions about the future research directions.
Collapse
|
47
|
González-Pérez M, Camasão DB, Mantovani D, Alonso M, Rodríguez-Cabello JC. Biocasting of an elastin-like recombinamer and collagen bi-layered model of the tunica adventitia and external elastic lamina of the vascular wall. Biomater Sci 2021; 9:3860-3874. [PMID: 33890956 DOI: 10.1039/d0bm02197k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of techniques for fabricating vascular wall models will foster the development of preventive and therapeutic therapies for treating cardiovascular diseases. However, the physical and biological complexity of vascular tissue represents a major challenge, especially for the design and the production of off-the-shelf biomimetic vascular replicas. Herein, we report the development of a biocasting technique that can be used to replicate the tunica adventitia and the external elastic lamina of the vascular wall. Type I collagen embedded with neonatal human dermal fibroblast (HDFn) and an elastic click cross-linkable, cell-adhesive and protease-sensitive elastin-like recombinamer (ELR) hydrogel were investigated as readily accessible and tunable layers to the envisaged model. Mechanical characterization confirmed that the viscous and elastic attributes predominated in the collagen and ELR layers, respectively. In vitro maturation confirmed that the collagen and ELR provided a favorable environment for the HDFn viability, while histology revealed the wavy and homogenous morphology of the ELR and collagen layer respectively, the cell polarization towards the cell-attachment sites encoded on the ELR, and the enhanced expression of glycosaminoglycan-rich extracellular matrix and differentiation of the embedded HDFn into myofibroblasts. As a complementary assay, 30% by weight of the collagen layer was substituted with the ELR. This model proved the possibility to tune the composition and confirm the versatile character of the technology developed, while revealing no significant differences with respect to the original construct. On-demand modification of the model dimensions, number and composition of the layers, as well as the type and density of the seeded cells, can be further envisioned, thus suggesting that this bi-layered model may be a promising platform for the fabrication of biomimetic vascular wall models.
Collapse
Affiliation(s)
- Miguel González-Pérez
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, 47011 Valladolid, Spain.
| | - Dimitria Bonizol Camasão
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Québec, Division of Regenerative Medicine, Laval University, Québec, QC, Canada G1V 0A6
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Québec, Division of Regenerative Medicine, Laval University, Québec, QC, Canada G1V 0A6
| | - Matilde Alonso
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, 47011 Valladolid, Spain.
| | - José Carlos Rodríguez-Cabello
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, 47011 Valladolid, Spain.
| |
Collapse
|
48
|
Chen EP, Toksoy Z, Davis BA, Geibel JP. 3D Bioprinting of Vascularized Tissues for in vitro and in vivo Applications. Front Bioeng Biotechnol 2021; 9:664188. [PMID: 34055761 PMCID: PMC8158943 DOI: 10.3389/fbioe.2021.664188] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/06/2021] [Indexed: 12/23/2022] Open
Abstract
With a limited supply of organ donors and available organs for transplantation, the aim of tissue engineering with three-dimensional (3D) bioprinting technology is to construct fully functional and viable tissue and organ replacements for various clinical applications. 3D bioprinting allows for the customization of complex tissue architecture with numerous combinations of materials and printing methods to build different tissue types, and eventually fully functional replacement organs. The main challenge of maintaining 3D printed tissue viability is the inclusion of complex vascular networks for nutrient transport and waste disposal. Rapid development and discoveries in recent years have taken huge strides toward perfecting the incorporation of vascular networks in 3D printed tissue and organs. In this review, we will discuss the latest advancements in fabricating vascularized tissue and organs including novel strategies and materials, and their applications. Our discussion will begin with the exploration of printing vasculature, progress through the current statuses of bioprinting tissue/organoids from bone to muscles to organs, and conclude with relevant applications for in vitro models and drug testing. We will also explore and discuss the current limitations of vascularized tissue engineering and some of the promising future directions this technology may bring.
Collapse
Affiliation(s)
- Earnest P Chen
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Yale College, Yale University, New Haven, CT, United States
| | - Zeren Toksoy
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Yale College, Yale University, New Haven, CT, United States
| | - Bruce A Davis
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT, United States
| | - John P Geibel
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
49
|
Deprez J, Lajoinie G, Engelen Y, De Smedt SC, Lentacker I. Opening doors with ultrasound and microbubbles: Beating biological barriers to promote drug delivery. Adv Drug Deliv Rev 2021; 172:9-36. [PMID: 33705877 DOI: 10.1016/j.addr.2021.02.015] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Apart from its clinical use in imaging, ultrasound has been thoroughly investigated as a tool to enhance drug delivery in a wide variety of applications. Therapeutic ultrasound, as such or combined with cavitating nuclei or microbubbles, has been explored to cross or permeabilize different biological barriers. This ability to access otherwise impermeable tissues in the body makes the combination of ultrasound and therapeutics very appealing to enhance drug delivery in situ. This review gives an overview of the most important biological barriers that can be tackled using ultrasound and aims to provide insight on how ultrasound has shown to improve accessibility as well as the biggest hurdles. In addition, we discuss the clinical applicability of therapeutic ultrasound with respect to the main challenges that must be addressed to enable the further progression of therapeutic ultrasound towards an effective, safe and easy-to-use treatment tailored for drug delivery in patients.
Collapse
Affiliation(s)
- J Deprez
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, P.O. Box 217, 7500 AE Enschede, Netherlands
| | - Y Engelen
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - S C De Smedt
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
50
|
Zhang YF, Zhang Y, Jia DD, Yang HY, Cheng MD, Zhu WX, Xin H, Li PF, Zhang YF. Insights into the regulatory role of Plexin D1 signalling in cardiovascular development and diseases. J Cell Mol Med 2021; 25:4183-4194. [PMID: 33837646 PMCID: PMC8093976 DOI: 10.1111/jcmm.16509] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/04/2021] [Accepted: 03/22/2021] [Indexed: 12/30/2022] Open
Abstract
Plexin D1 (PLXND1), which was previously thought to mediate semaphorin signalling, belongs to the Plexin family of transmembrane proteins. PLXND1 cooperates mostly with the coreceptor neuropilin and participates in many aspects of axonal guidance. PLXND1 can also act as both a tumour promoter and a tumour suppressor. Emerging evidence suggests that mutations in PLXND1 or Semaphorin 3E, the canonical ligand of PLXND1, can lead to serious cardiovascular diseases, such as congenital heart defects, CHARGE syndrome and systemic sclerosis. Upon ligand binding, PLXND1 can act as a GTPase‐activating protein (GAP) and modulate integrin‐mediated cell adhesion, cytoskeletal dynamics and cell migration. These effects may play regulatory roles in the development of the cardiovascular system and disease. The cardiovascular effects of PLXND1 signalling have gradually been elucidated. PLXND1 was recently shown to detect physical forces and translate them into intracellular biochemical signals in the context of atherosclerosis. Therefore, the role of PLXND1 in cardiovascular development and diseases is gaining research interest because of its potential as a biomarker and therapeutic target. In this review, we describe the cardiac effects, vascular effects and possible molecular mechanisms of PLXND1 signalling.
Collapse
Affiliation(s)
- Yi-Fei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Dong-Dong Jia
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Hong-Yu Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Meng-Die Cheng
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wen-Xiu Zhu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Pei-Feng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|