1
|
Kopacz A, Kloska D, Bar A, Targosz-Korecka M, Cysewski D, Awsiuk K, Piechota-Polanczyk A, Cichon M, Chlopicki S, Jozkowicz A, Grochot-Przeczek A. Endothelial miR-34a deletion guards against aneurysm development despite endothelial dysfunction. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167812. [PMID: 40139409 DOI: 10.1016/j.bbadis.2025.167812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
We previously reported a link between NRF2, a cytoprotective transcription factor, and the ageing of endothelial cells (ECs) and aorta. We also found that NRF2 KO mice are more susceptible to the development of abdominal aortic aneurysm (AAA), which is an age-associated condition. Since miR-34a is a marker of ageing, we explored its relationship with NRF2 and its role in vascular function and AAA formation. Here, we demonstrate that premature NRF2-dependent ageing of ECs is mediated by miR-34a. Infusion of hypertensive angiotensin II (Ang II) in mice increases miR-34a in the aortic endothelial layer and serum, particularly in mice developing AAA. Mice lacking endothelial miR-34a exhibit severe EC dysfunction. Despite that, they are protected from AAA, also on the NRF2 KO background. This protective effect is reversed by rapamycin, which suppresses Ang II-induced EC proliferation. We identified MTA2, but not SIRT1, as a target of miR-34a that inhibits EC proliferation stimulated by Ang II. These findings suggest that fine-tuning of EC proliferation could have potential therapeutic implications for the treatment of aneurysms.
Collapse
Affiliation(s)
- Aleksandra Kopacz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Damian Kloska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Marta Targosz-Korecka
- Department of Physics of Nanostructures and Nanotechnology, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Krakow, Poland
| | - Dominik Cysewski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Kamil Awsiuk
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Krakow, Poland
| | - Aleksandra Piechota-Polanczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Milena Cichon
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
2
|
Gomez-Salinero JM, Redmond D, Rafii S. Microenvironmental determinants of endothelial cell heterogeneity. Nat Rev Mol Cell Biol 2025; 26:476-495. [PMID: 39875728 DOI: 10.1038/s41580-024-00825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
During development, endothelial cells (ECs) undergo an extraordinary specialization by which generic capillary microcirculatory networks spanning from arteries to veins transform into patterned organotypic zonated blood vessels. These capillary ECs become specialized to support the cellular and metabolic demands of each specific organ, including supplying tissue-specific angiocrine factors that orchestrate organ development, maintenance of organ-specific functions and regeneration of injured adult organs. Here, we illustrate the mechanisms by which microenvironmental signals emanating from non-vascular niche cells induce generic ECs to acquire specific inter-organ and intra-organ functional attributes. We describe how perivascular, parenchymal and immune cells dictate vascular heterogeneity and capillary zonation, and how this system is maintained through tissue-specific signalling activated by vasculogenic and angiogenic factors and deposition of matrix components. We also discuss how perturbation of organotypic vascular niche cues lead to erasure of EC signatures, contributing to the pathogenesis of disease processes. We also describe approaches that use reconstitution of tissue-specific signatures of ECs to promote regeneration of damaged organs.
Collapse
Affiliation(s)
- Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
3
|
Ghosh S, Bishnoi B, Das S. Artery regeneration: Molecules, mechanisms and impact on organ function. Semin Cell Dev Biol 2025; 171:103611. [PMID: 40318557 DOI: 10.1016/j.semcdb.2025.103611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 05/07/2025]
Abstract
Replenishment of artery cells to repair or create new arteries is a promising strategy to re-vascularize ischemic tissue. However, limited understanding of cellular and molecular programs associated with artery (re-)growth impedes our efforts towards designing optimal therapeutic approaches. In this review, we summarize different cellular mechanisms that drive injury-induced artery regeneration in distinct organs and organisms. Artery formation during embryogenesis includes migration, self-amplification, and changes in cell fates. These processes are coordinated by multiple signaling pathways, like Vegf, Wnt, Notch, Cxcr4; many of which, also involved in injury-induced vascular responses. We also highlight how physiological and environmental factors determine the extent of arterial re-vascularization. Finally, we discuss different in vitro cellular reprogramming and tissue engineering approaches to promote artery regeneration, in vivo. This review provides the current understanding of endothelial cell fate reprogramming and explores avenues for regenerating arteries to restore organ function through efficient revascularization.
Collapse
Affiliation(s)
- Swarnadip Ghosh
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Bhavnesh Bishnoi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Soumyashree Das
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India.
| |
Collapse
|
4
|
Fazzari J, Fernandez-Palomo C, Pellicioli P, Day L, Trappetti V, Lucien-Matteoni F, Kim Y, Mutter R, Park S, Grams M, Djonov V. Spatially fractionated minibeam radiation delivered at clinically feasible dose rates induces transient vascular permeability. Sci Rep 2025; 15:8210. [PMID: 40064939 PMCID: PMC11894116 DOI: 10.1038/s41598-025-87395-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/20/2025] [Indexed: 03/14/2025] Open
Abstract
Microbeam Radiation Therapy is a preclinical form of spatially fractionated radiation therapy that utilizes synchrotron X-rays to deliver highly heterogeneous dose distributions at a micrometric scale. This radiation scheme has been shown to facilitate the induction of controlled and reversible vascular permeability, enhancing treatment efficacy of systemic therapeutic agents. Despite the promising preclinical results, translating microbeam SFRT to the clinic has been hindered by a reliance on synchrotron sources that operate at dose rates orders of magnitude greater than what is possible with clinical machines. Without rapid dose delivery, the microbeam geometry is susceptible to blurring due to physiologic motion when delivered at clinical dose rates. Therefore, larger beam widths, spaced further apart (minibeams) were employed to determine whether such effects can be observed with clinically achievable doses and dose rates. Vascular permeability was assessed in the chick chorioallantoic membrane vasculature following minibeam irradiation delivered at peak doses (10 Gy) and dose rates (10 Gy/s and 0.05 Gy/s) approaching clinical relevance. Transient, reversible permeability could be induced at these dose rates beginning 1-2 h post-irradiation. This was followed by temporary vascular occlusion in the beam path that was resolved by 7 h when delivered at 10 Gy/s but persisted longer when delivered at 0.05 Gy/s. Despite these changes, vascular function was maintained at both dose rates by 24 h post-IR, differing only in the degree of regeneration. The induction of permeability was also maintained when using a clinical orthovoltage system further supporting the potential clinical application of minibeam radiation therapy.
Collapse
Affiliation(s)
| | | | - Paolo Pellicioli
- Institute of Anatomy, University of Bern, Bern, Switzerland
- ID17 Biomedical Beamline, European Synchrotron Radiation Facility, Grenoble, France
| | - Liam Day
- Institute of Anatomy, University of Bern, Bern, Switzerland
- ID17 Biomedical Beamline, European Synchrotron Radiation Facility, Grenoble, France
| | | | | | - Yohan Kim
- Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - Robert Mutter
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
- Department of Pharmacology, Mayo Clinic, Rochester, MN, USA
| | - Sean Park
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Michael Grams
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Bern, Switzerland.
| |
Collapse
|
5
|
Xu X, Zhu P, Wang H, Chen K, Liu L, Du L, Jiang L, Hu Y, Zhou X, Zhang B, Pu X, Hu X, Xu Q, Zhang L, Li W. CD34 + PI16 + fibroblast progenitors aggravate neointimal lesions of allograft arteries via CCL11/CCR3-PI3K/AKT pathway. Theranostics 2025; 15:2523-2543. [PMID: 39990233 PMCID: PMC11840720 DOI: 10.7150/thno.104650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/01/2025] [Indexed: 02/25/2025] Open
Abstract
Rationale: Transplant-accelerated arteriosclerosis is a common complication that limits the long-term survival of organ transplant recipients. While previous studies have indicated the involvement of CD34+ stem/progenitor cells (SPCs) in this process, their heterogeneity and potential adverse effects remains incompletely understood. Methods: To investigate the role of CD34+ SPCs in transplant arteriosclerosis, we used various genetically modified mouse models, including BALB/c, C57BL/6J, CD34-CreERT2, Rosa26-tdTomato, Rosa26-iDTR, CD34-Dre, PI16-CreERT2, and CAG-LSL-RSR-tdTomato-2A-DTR mice. Single-cell RNA sequencing (scRNA-seq), chemokine antibody microarrays, ELISA assays, and immunohistochemistry were employed to identify fibroblast progenitors and their interactions with smooth muscle cells. Furthermore, in vivo and in vitro experiments targeting the CCL11/CCR3-PI3K/AKT signaling pathway were conducted to assess its role in the pathogenesis of transplant arteriosclerosis. Results: Single-cell RNA-seq and genetic lineage tracing revealed a subpopulation of fibroblast progenitors, characterized by high CD34 and PI16 expression, which differentiated into a distinct chemotactic fibroblast subset. Proteomic and scRNA analysis revealed that this CD34+ PI16- subgroup released CCL11 (Eotaxin-1), which promoted intimal hyperplasia through the paracrine activation of smooth muscle cells. Binding of CCL11 to its receptor CCR3 activated the PI3K/AKT signaling pathway in smooth muscle cells, driving their proliferation and migration. In vivo, overexpression of CCL11 promoted neointimal hyperplasia, while neutralizing CCL11 or inhibiting CCR3 alleviated neointimal formation. Conclusions: These findings identified CD34+ PI16+ fibroblast progenitors that differentiate into specific chemotactic fibroblasts, releasing chemokines pivotal for neointima formation, suggesting a therapeutic strategy targeting their chemotactic activity.
Collapse
Affiliation(s)
- Xiaodong Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengwei Zhu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Han Wang
- School of Engineering and Materials Science, Queen Mary University of London, United Kingdom
| | - Kai Chen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Liu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Luping Du
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuhao Zhou
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bohuan Zhang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangyuan Pu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaosheng Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Zhang
- Department of Cardiology, and Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weidong Li
- Department of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Sesorova IS, Bedyaev EV, Vavilov PS, Levin SL, Mironov AA. Regeneration of Vascular Endothelium in Different Large Vessels. Int J Mol Sci 2025; 26:837. [PMID: 39859550 PMCID: PMC11766047 DOI: 10.3390/ijms26020837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 01/27/2025] Open
Abstract
The regeneration of endothelial cells (ECs) lining arteries, veins, and large lymphatic vessels plays an important role in vascular pathology. To understand the mechanisms of atherogenesis, it is important to determine what happens during endothelial regeneration. A comparison of these processes in the above-mentioned vessels reveals both similarities and some significant differences. Regeneration is carried out by moving intact ECs from the edges of the viable endothelial layer towards the centre of the EC damage zone. A sharp decrease in contact inhibition leads to the spreading of the edges of the ECs situated on the damage border. This stimulates the second row of ECs to enter the S-phase, then the G2 phase of cell cycle, and finally mitosis. In all three types of vessels studied, mitotically dividing ECs were found using correlation light and electron microscopy. These ECs have a body protruding into the lumen of the vessel, covered with micro-villi and other outgrowths. The level of EC rounding and protruding is highest in the arteries and least pronounced in the lymphatic vessels. The intercellular contacts of mitotically dividing cells become wider. The EC division leads to an increase in the density of ECs. ECs moving over the damaged area and partially outside the damaged area acquire a fusiform shape. In the process of regeneration of arterial endothelium, the damaged ECs are removed. Then health ECs move to a surface devoid of endothelium, and detach spreading out, flattened platelets from the luminal surface of the vessel. In the veins, ECs grow on the surface of platelets and microthrombi. In lymphatic vessels, ECs detach from the basement membrane slower than in the veins and arteries. There, the migrating ECs grow under fibrin fibres. After some time (usually after 30 days), the EC mosaic returns to normal in all three types of vessels.
Collapse
Affiliation(s)
- Irina S. Sesorova
- Department of Anatomy, Ivanovo State Medical University, 153012 Ivanovo, Russia; (I.S.S.); (E.V.B.); (P.S.V.)
| | - Eugeny V. Bedyaev
- Department of Anatomy, Ivanovo State Medical University, 153012 Ivanovo, Russia; (I.S.S.); (E.V.B.); (P.S.V.)
| | - Pavel S. Vavilov
- Department of Anatomy, Ivanovo State Medical University, 153012 Ivanovo, Russia; (I.S.S.); (E.V.B.); (P.S.V.)
| | - Sergei L. Levin
- The EMC European Medical Centre, Central JSC RU, Spiridonyevsky Lane, 5, 123001 Moscow, Russia;
| | - Alexander A. Mironov
- Department of Cell Biology, IFOM ETS—The AIRC Institute of Molecular Oncology, Via Adamello, 16, 20139 Milan, Italy
| |
Collapse
|
7
|
Cantalupo A, Asano K, Dikalov S, Gordon D, Ramirez F. Fibrillin-1 Deficiency Perturbs Aortic Cholinergic Relaxation and Adrenergic Contraction in a Mouse Model of Early Onset Progressively Severe Marfan Syndrome. J Vasc Res 2025; 62:96-108. [PMID: 39827863 PMCID: PMC11961321 DOI: 10.1159/000542481] [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: 05/28/2024] [Accepted: 11/03/2024] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION The pathogenic role of nitric oxide (NO) signaling during development of thoracic aortic aneurysm (TAA) in Marfan syndrome (MFS) is currently unclear. We characterized vasomotor function and its relationship to the activity of the NO-generating enzymes in mice with early onset progressively severe MFS. METHODS Wire myography, immunoblotting, measurements of aortic NO, and superoxide levels were used to compare vasomotor function, contractile protein levels, and the activity of endothelial and inducible NO synthase (eNOS and iNOS, respectively) in ascending thoracic aortas of Fbn1mgR/mgR mice relative to wild-type littermates. RESULTS Isometric force measurements of aortic rings from 16-day-old male Fbn1mgR/mgR mice revealed a significant reduction in acetylcholine-induced relaxation and increased phenylephrine (PE)-promoted contractility, associated with abnormally low eNOSSer1177 phosphorylation, decreased NO production, and augmented superoxide levels. Greater aortic contractility was associated with α1-adrenoceptor upregulation and normal levels of contractile proteins. While iNOS inhibition had no effect on vasomotor functions, mutant aortic rings preincubated with a nonspecific NOS inhibitor yielded a greater PE response, implying a significant contribution of endothelial dysfunction to aortic hypercontractility. CONCLUSION Impaired eNOS signaling disrupts aortic cholinergic relaxation and adrenergic contraction in MFS mice with dissecting TAA. INTRODUCTION The pathogenic role of nitric oxide (NO) signaling during development of thoracic aortic aneurysm (TAA) in Marfan syndrome (MFS) is currently unclear. We characterized vasomotor function and its relationship to the activity of the NO-generating enzymes in mice with early onset progressively severe MFS. METHODS Wire myography, immunoblotting, measurements of aortic NO, and superoxide levels were used to compare vasomotor function, contractile protein levels, and the activity of endothelial and inducible NO synthase (eNOS and iNOS, respectively) in ascending thoracic aortas of Fbn1mgR/mgR mice relative to wild-type littermates. RESULTS Isometric force measurements of aortic rings from 16-day-old male Fbn1mgR/mgR mice revealed a significant reduction in acetylcholine-induced relaxation and increased phenylephrine (PE)-promoted contractility, associated with abnormally low eNOSSer1177 phosphorylation, decreased NO production, and augmented superoxide levels. Greater aortic contractility was associated with α1-adrenoceptor upregulation and normal levels of contractile proteins. While iNOS inhibition had no effect on vasomotor functions, mutant aortic rings preincubated with a nonspecific NOS inhibitor yielded a greater PE response, implying a significant contribution of endothelial dysfunction to aortic hypercontractility. CONCLUSION Impaired eNOS signaling disrupts aortic cholinergic relaxation and adrenergic contraction in MFS mice with dissecting TAA.
Collapse
MESH Headings
- Animals
- Marfan Syndrome/physiopathology
- Marfan Syndrome/genetics
- Marfan Syndrome/metabolism
- Marfan Syndrome/complications
- Fibrillin-1/deficiency
- Fibrillin-1/genetics
- Male
- Disease Models, Animal
- Nitric Oxide Synthase Type III/metabolism
- Aorta, Thoracic/physiopathology
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/drug effects
- Nitric Oxide/metabolism
- Vasodilation/drug effects
- Vasoconstriction/drug effects
- Nitric Oxide Synthase Type II/metabolism
- Aortic Aneurysm, Thoracic/physiopathology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/etiology
- Acetylcholine/pharmacology
- Superoxides/metabolism
- Receptors, Adrenergic, alpha-1/metabolism
- Receptors, Adrenergic, alpha-1/drug effects
- Signal Transduction
- Phosphorylation
- Mice, Inbred C57BL
- Adrenergic alpha-1 Receptor Agonists/pharmacology
- Severity of Illness Index
- Phenylephrine/pharmacology
- Mice
- Vasodilator Agents/pharmacology
- Disease Progression
- Adipokines
Collapse
Affiliation(s)
- Anna Cantalupo
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keiichi Asano
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergey Dikalov
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dylan Gordon
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francesco Ramirez
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
8
|
Ueda C, Sakimoto S, Yoshihara M, Takigawa T, Shiraki A, Yamaguchi K, Shiki K, Shiraki N, Kitajima S, Kubota Y, Fukushima Y, Nishida K. Endothelial activating transcription factor 3 promotes angiogenesis and vascular repair in the mouse retina. iScience 2025; 28:111516. [PMID: 39790557 PMCID: PMC11714383 DOI: 10.1016/j.isci.2024.111516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 06/26/2024] [Accepted: 11/08/2024] [Indexed: 01/12/2025] Open
Abstract
Ischemia and pathological angiogenesis in retinal vascular diseases cause serious vision-related problems. However, the transcriptional regulators of vascular repair remain unidentified. Thus, the factors and mechanisms involved in angiogenesis must be elucidated to develop approaches for restoring normal blood vessels. Here, we investigated the effects of the stress response activating transcription factor 3 (ATF3) on angiogenesis and vascular regeneration in vitro and in vivo. ATF3 was expressed specifically in retinal vascular endothelial cells (ECs) during vascular development. Vascular endothelial growth factor stimulation upregulated ATF3 expression in cultured ECs. The downregulated ATF3 expression in ECs caused the deterioration of vascular network formation in vitro and in vivo. Moreover, ATF3 deletion in a model of oxygen-induced retinopathy inhibited retinal vascular repair but not pathological neovascularization. Transcriptome analysis confirmed that high ATF3 expression upregulated the expression of angiogenesis-related genes in ECs. ATF3 may aid vascular repair therapy in retinal vascular diseases.
Collapse
Affiliation(s)
- Chihiro Ueda
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Susumu Sakimoto
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Masahito Yoshihara
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toru Takigawa
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akihiko Shiraki
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kaito Yamaguchi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kosuke Shiki
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Nobuhiko Shiraki
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shigetaka Kitajima
- Department of Biochemical Genetics, Medical Research Institute and Laboratory of Genome Structure and Regulation, School of Biomedical Science, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Yoko Fukushima
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka, Japan
| |
Collapse
|
9
|
Pi D, Braun J, Dutta S, Patra D, Bougaran P, Mompeón A, Ma F, Stock SR, Choi S, García-Ortega L, Pratama MY, Pichardo D, Ramkhelawon B, Benedito R, Bautch VL, Ornitz DM, Goyal Y, Iruela-Arispe ML. Resolving the design principles that control postnatal vascular growth and scaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627758. [PMID: 39713449 PMCID: PMC11661209 DOI: 10.1101/2024.12.10.627758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
After birth, tissues grow continuously until reaching adult size, with each organ exhibiting unique cellular dynamics, growth patterns, and (stem or non-stem) cell sources. Using a suite of experimental and computational multiscale approaches, we found that aortic expansion is guided by specific biological principles and scales with the vertebral column rather than animal body weight. Expansion proceeds via two distinct waves of arterial cell proliferation along blood flow that are spatially stochastic, yet temporally coordinated. Each wave exhibits unique cell cycle kinetics and properties, with the first wave exhibiting cell cycle durations as fast as 6 hours. Single-cell RNA sequencing showed changes in fatty acid metabolism concomitant with an increase in cell size. Mathematical modeling and experiments indicated endothelial cell extrusion is essential for homeostatic aortic growth and balancing excess proliferation. In a genetic model of achondroplasia, the aorta achieves proper scaling through enhanced cell extrusion while maintaining normal proliferation dynamics. Collectively, these results provide a blueprint of the principles that orchestrate aortic growth which depends entirely on differentiated cell proliferation rather than resident stem cells.
Collapse
|
10
|
Huveneers S, Phng LK. Endothelial cell mechanics and dynamics in angiogenesis. Curr Opin Cell Biol 2024; 91:102441. [PMID: 39342870 DOI: 10.1016/j.ceb.2024.102441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024]
Abstract
The efficient distribution of oxygen and metabolites is critical for embryonic development and growth as well as tissue homeostasis. This is achieved by endothelial cells forming and maintaining a closed, circulatory network of tubular blood vessels. Endothelial cells are highly plastic cells with the capability to generate diverse dynamic responses at different stages of vessel development in order to build vessel networks of tissue-specific patterns and morphologies. In this review, we discuss new conceptual advances gained from in vitro and in vivo models of angiogenesis on the control of endothelial cell dynamics. We highlight the complex interplay between mechanical cues, actin cytoskeleton and endothelial behaviors, and the emerging importance of hydrostatic pressure in complementing actin-dependent mechanisms to regulate endothelial cell mechanics and angiogenesis. Understanding these processes provides insights into vascular repair and regeneration mechanisms.
Collapse
Affiliation(s)
- Stephan Huveneers
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
| | - Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.
| |
Collapse
|
11
|
Skulimowska I, Morys J, Sosniak J, Gonka M, Gulati G, Sinha R, Kowalski K, Mosiolek S, Weissman IL, Jozkowicz A, Szade A, Szade K. Polyclonal regeneration of mouse bone marrow endothelial cells after irradiative conditioning. Cell Rep 2024; 43:114779. [PMID: 39489938 PMCID: PMC11602546 DOI: 10.1016/j.celrep.2024.114779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 06/04/2024] [Accepted: 09/04/2024] [Indexed: 11/05/2024] Open
Abstract
Bone marrow endothelial cells (BM-ECs) are the essential components of the BM niche and support the function of hematopoietic stem cells (HSCs). However, conditioning for HSC transplantation causes damage to the recipients' BM-ECs and may lead to transplantation-related morbidity. Here, we investigated the cellular and clonal mechanisms of BM-EC regeneration after irradiative conditioning. Using single-cell RNA sequencing, imaging, and flow cytometry, we revealed how the heterogeneous pool of BM-ECs changes during regeneration from irradiation stress. Next, we developed a single-cell in vitro clonogenic assay and demonstrated that all EC fractions hold a high potential to reenter the cell cycle and form vessel-like structures. Finally, we used Rainbow mice and a machine-learning-based model to show that the regeneration of BM-ECs after irradiation is mostly polyclonal and driven by the broad fraction of BM-ECs; however, the cell output among clones varies at later stages of regeneration.
Collapse
Affiliation(s)
- Izabella Skulimowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland; Laboratory of Stem Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland; Doctoral School of Exact and Natural Sciences, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Jan Morys
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Justyna Sosniak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Monika Gonka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Gunsagar Gulati
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kacper Kowalski
- Laboratory of Stem Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Sylwester Mosiolek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Agata Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Krzysztof Szade
- Laboratory of Stem Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.
| |
Collapse
|
12
|
Moise K, Arun KM, Pillai M, Salvador J, Mehta AS, Goyal Y, Iruela-Arispe ML. Endothelial cell elongation and alignment in response to shear stress requires acetylation of microtubules. Front Physiol 2024; 15:1425620. [PMID: 39318362 PMCID: PMC11420013 DOI: 10.3389/fphys.2024.1425620] [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: 04/30/2024] [Accepted: 08/20/2024] [Indexed: 09/26/2024] Open
Abstract
The innermost layer of the vessel wall is constantly subjected to recurring and relenting mechanical forces by virtue of their direct contact with blood flow. Endothelial cells of the vessel are exposed to distension, pressure, and shear stress; adaptation to these hemodynamic forces requires significant remodeling of the cytoskeleton which includes changes in actin, intermediate filaments, and microtubules. While much is known about the effect of shear stress on the endothelial actin cytoskeleton; the impact of hemodynamic forces on the microtubule network has not been investigated in depth. Here we used imaging techniques and protein expression analysis to characterize how pharmacological and genetic perturbations of microtubule properties alter endothelial responses to laminar shear stress. Our findings revealed that pharmacological suppression of microtubule dynamics blocked two typical responses to laminar shear stress: endothelial elongation and alignment. The findings demonstrate the essential contribution of the microtubule network to changes in cell shape driven by mechanical forces. Furthermore, we observed a flow-dependent increase in microtubule acetylation that occurred early in the process of cell elongation. Pharmacological manipulation of microtubule acetylation showed a direct and causal relationship between acetylation and endothelial elongation. Finally, genetic inactivation of aTAT1, a microtubule acetylase, led to significant loss of acetylation as well as inhibition of cell elongation in response to flow. In contrast, loss of HDAC6, a microtubule deacetylase, resulted in robust microtubule acetylation with cells displaying faster kinetics of elongation and alignment. Taken together, our findings uncovered the critical contributions of HDAC6 and aTAT1, that through their roles in the regulation of microtubule acetylation, are key mediators of endothelial mechanotransduction.
Collapse
Affiliation(s)
- Katiannah Moise
- Department of Cell and Development Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| | - Keerthana M. Arun
- Center for Synthetic Biology, Northwestern University, Chicago, IL, United States
| | - Maalavika Pillai
- Department of Cell and Development Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
- Center for Synthetic Biology, Northwestern University, Chicago, IL, United States
| | - Jocelynda Salvador
- Department of Cell and Development Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| | - Aarya S. Mehta
- Department of Cell and Development Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| | - Yogesh Goyal
- Department of Cell and Development Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
- Center for Synthetic Biology, Northwestern University, Chicago, IL, United States
| | - M. Luisa Iruela-Arispe
- Department of Cell and Development Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
13
|
Xu J, Wang J, Zhang H, Chen Y, Zhang X, Zhang Y, Xie M, Xiao J, Qiu J, Wang G. Coupled single-cell and bulk RNA-seq analysis reveals the engulfment role of endothelial cells in atherosclerosis. Genes Dis 2024; 11:101250. [PMID: 39022128 PMCID: PMC11252887 DOI: 10.1016/j.gendis.2024.101250] [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: 12/12/2022] [Revised: 10/28/2023] [Accepted: 12/05/2023] [Indexed: 07/20/2024] Open
Abstract
The clearance of apoptotic cell debris, containing professional phagocytosis and non-professional phagocytosis, is essential for maintaining the homeostasis of healthy tissues. Here, we discovered that endothelial cells could engulf apoptotic cell debris in atherosclerotic plaque. Single-cell RNA sequencing (RNA-seq) has revealed a unique endothelial cell subpopulation in atherosclerosis, which was strongly associated with vascular injury-related pathways. Moreover, integrated analysis of three vascular injury-related RNA-seq datasets showed that the expression of scavenger receptor class B type 1 (SR-B1) was up-regulated and specifically enriched in the phagocytosis pathway under vascular injury circumstances. Single-cell RNA-seq and bulk RNA-seq indicate that SR-B1 was highly expressed in a unique endothelial cell subpopulation of mouse aorta and strongly associated with the reorganization of cellular adherent junctions and cytoskeleton which were necessary for phagocytosis. Furthermore, SR-B1 was strongly required for endothelial cells to engulf apoptotic cell debris in atherosclerotic plaque of both mouse and human aorta. Overall, this study demonstrated that apoptotic cell debris could be engulfed by endothelial cells through SR-B1 and associated with the reorganization of cellular adherent junctions and cytoskeleton.
Collapse
Affiliation(s)
- Jianxiong Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Jinxuan Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Hongping Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Yidan Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Xiaojuan Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Ying Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
- Chongqing Emergency Medical Center (Chongqing University Central Hospital), Chongqing 400014, China
| | - Ming Xie
- Chongqing Emergency Medical Center (Chongqing University Central Hospital), Chongqing 400014, China
| | - Jun Xiao
- Chongqing Emergency Medical Center (Chongqing University Central Hospital), Chongqing 400014, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
14
|
Li K, Dai M, Sacirovic M, Pagonas N, Ritter O, Kah J, Lauxmann MA, Bramlage P, Bondke Persson A, Buschmann I, Hillmeister P. Association of endothelial function and lower extremity perfusion in peripheral artery disease. VASA 2024; 53:333-340. [PMID: 38979892 DOI: 10.1024/0301-1526/a001131] [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: 07/10/2024]
Abstract
Background: The current study aims to investigate the association between endothelial function and lower extremity perfusion in patients with peripheral artery disease (PAD). Patients and methods: In total 229 patients with PAD (Rutherford stage 0-3) were enrolled in the current study. Endothelial function was assessed by measuring flow-mediated dilation (FMD) and endothelial cell proliferation capacity (ECPC). Lower extremity perfusion was assessed by measuring oscillometry-based ankle brachial index (oABI) and pulse wave index (PWI). In addition, carotid intima-media-thickness (cIMT) was also measured as a surrogate marker for atherosclerosis. Correlations between FMD, ECPC, oABI, PWI, and cIMT were analysed using Pearson correlation coefficient. The relationship between the above variables and the severity of PAD was investigated using ordinal logistic regression analysis. Results: Correlation analysis showed that FMD negatively associated with PWI (r = -0.183, p = 0.005), ECPC positively associated with oABI (r = 0.162, p = 0.014), and oABI negatively associated with PWI (r = -0.264, p < 0.001). Ordinal logistic regression analysis showed that ECPC (β = -0.009, p = 0.048), oABI (β = -5.290, p < 0.001), and age (β = -0.058, p = 0.002) negatively associated with the PAD Rutherford stages. In addition, PWI (β = 0.006, p < 0.001), cIMT (β = 18.363, p = 0.043) positively associated with the PAD Rutherford stages. Conclusions: Endothelial function significantly associates with lower extremity perfusion in patients with PAD, and both are related to the severity of PAD.
Collapse
Affiliation(s)
- Kangbo Li
- Department of Angiology, Center for Internal Medicine I, Deutsches Angiologie-Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mengjun Dai
- Department of Angiology, Center for Internal Medicine I, Deutsches Angiologie-Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mesud Sacirovic
- Department of Angiology, Center for Internal Medicine I, Deutsches Angiologie-Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Nikolaos Pagonas
- Department of Cardiology, Center for Internal Medicine I, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Oliver Ritter
- Department of Cardiology, Center for Internal Medicine I, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Janine Kah
- Department of Gastroenterology, Center for Internal Medicine II, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Martin A Lauxmann
- Institute of Biochemistry, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Peter Bramlage
- Institute for Pharmacology and Preventive Medicine, Cloppenburg, Germany
| | - Anja Bondke Persson
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ivo Buschmann
- Department of Angiology, Center for Internal Medicine I, Deutsches Angiologie-Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Philipp Hillmeister
- Department of Angiology, Center for Internal Medicine I, Deutsches Angiologie-Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| |
Collapse
|
15
|
Jeong JY, Bafor AE, Freeman BH, Chen PR, Park ES, Kim E. Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines 2024; 12:1795. [PMID: 39200259 PMCID: PMC11351371 DOI: 10.3390/biomedicines12081795] [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: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Brain arteriovenous malformations (bAVMs) substantially increase the risk for intracerebral hemorrhage (ICH), which is associated with significant morbidity and mortality. However, the treatment options for bAVMs are severely limited, primarily relying on invasive methods that carry their own risks for intraoperative hemorrhage or even death. Currently, there are no pharmaceutical agents shown to treat this condition, primarily due to a poor understanding of bAVM pathophysiology. For the last decade, bAVM research has made significant advances, including the identification of novel genetic mutations and relevant signaling in bAVM development. However, bAVM pathophysiology is still largely unclear. Further investigation is required to understand the detailed cellular and molecular mechanisms involved, which will enable the development of safer and more effective treatment options. Endothelial cells (ECs), the cells that line the vascular lumen, are integral to the pathogenesis of bAVMs. Understanding the fundamental role of ECs in pathological conditions is crucial to unraveling bAVM pathophysiology. This review focuses on the current knowledge of bAVM-relevant signaling pathways and dysfunctions in ECs, particularly the endothelial-to-mesenchymal transition (EndMT).
Collapse
Affiliation(s)
| | | | | | | | | | - Eunhee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.Y.J.); (A.E.B.); (B.H.F.); (P.R.C.); (E.S.P.)
| |
Collapse
|
16
|
Zhao J, Sormani L, Jacquelin S, Li H, Styke C, Zhou C, Beesley J, Oon L, Kaur S, Sim SL, Wong HY, Dight J, Hashemi G, Shafiee A, Roy E, Patel J, Khosrotehrani K. Distinct roles of SOX9 in self-renewal of progenitors and mesenchymal transition of the endothelium. Angiogenesis 2024; 27:545-560. [PMID: 38733496 PMCID: PMC11303482 DOI: 10.1007/s10456-024-09927-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/24/2024] [Indexed: 05/13/2024]
Abstract
Regenerative capabilities of the endothelium rely on vessel-resident progenitors termed endothelial colony forming cells (ECFCs). This study aimed to investigate if these progenitors are impacted by conditions (i.e., obesity or atherosclerosis) characterized by increased serum levels of oxidized low-density lipoprotein (oxLDL), a known inducer of Endothelial-to-Mesenchymal Transition (EndMT). Our investigation focused on understanding the effects of EndMT on the self-renewal capabilities of progenitors and the associated molecular alterations. In the presence of oxLDL, ECFCs displayed classical features of EndMT, through reduced endothelial gene and protein expression, function as well as increased mesenchymal genes, contractility, and motility. Additionally, ECFCs displayed a dramatic loss in self-renewal capacity in the presence of oxLDL. RNA-sequencing analysis of ECFCs exposed to oxLDL validated gene expression changes suggesting EndMT and identified SOX9 as one of the highly differentially expressed genes. ATAC sequencing analysis identified SOX9 binding sites associated with regions of dynamic chromosome accessibility resulting from oxLDL exposure, further pointing to its importance. EndMT phenotype and gene expression changes induced by oxLDL in vitro or high fat diet (HFD) in vivo were reversed by the silencing of SOX9 in ECFCs or the endothelial-specific conditional knockout of Sox9 in murine models. Overall, our findings support that EndMT affects vessel-resident endothelial progenitor's self-renewal. SOX9 activation is an early transcriptional event that drives the mesenchymal transition of endothelial progenitor cells. The identification of the molecular network driving EndMT in vessel-resident endothelial progenitors presents a new avenue in understanding and preventing a range of condition where this process is involved.
Collapse
Affiliation(s)
- Jilai Zhao
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Laura Sormani
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Sebastien Jacquelin
- Mater Research, Translational Research Institute, Macrophage Biology Laboratory, Brisbane, QLD, 4102, Australia
| | - Haiming Li
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Cassandra Styke
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Chenhao Zhou
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Jonathan Beesley
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Linus Oon
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Simranpreet Kaur
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
- Mater Research, Translational Research Institute, Macrophage Biology Laboratory, Brisbane, QLD, 4102, Australia
| | - Seen-Ling Sim
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Ho Yi Wong
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - James Dight
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Ghazaleh Hashemi
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Abbas Shafiee
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Edwige Roy
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia
| | - Jatin Patel
- Centre for Ageing Research Program, Queensland University of Technology, Brisbane, QLD, 4102, Australia
| | - Kiarash Khosrotehrani
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Experimental Dermatology Group, Brisbane, QLD, 4102, Australia.
| |
Collapse
|
17
|
Basuthakur P, Roy A, Ghosh S, Vijay V, Sinha D, Radhakrishnan M, Kumar A, Patra CR, Chakravarty S. Pro-angiogenic Terbium Hydroxide Nanorods Improve Critical Limb Ischemia in Part by Amelioration of Ischemia-Induced Endothelial Injury. ACS APPLIED BIO MATERIALS 2024; 7:4389-4405. [PMID: 38848346 DOI: 10.1021/acsabm.4c00252] [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: 06/09/2024]
Abstract
Critical limb ischemia (CLI) refers to a severe condition resulting from gradual obstruction in the supply of blood, oxygen, and nutrients to the limbs. The most promising clinical solution to CLI is therapeutic angiogenesis. This study explored the potency of pro-angiogenic terbium hydroxide nanorods (THNR) for treatment of CLI, with a major focus on their impact on ischemia-induced maladaptive alterations in endothelial cells as well as on vascularization in ischemic limbs. This study demonstrated that, in hypoxia-exposed endothelial cells, THNR improve survival and promote proliferation, migration, restoration of nitric oxide production, and regulation of vascular permeability. Based on molecular studies, these attributes of THNR can be traced to the stimulation of PI3K/AKT/eNOS signaling pathways. Besides, Wnt/GSK-3β/β-catenin signaling pathways may also play a role in the therapeutic actions of THNR. Furthermore, in the murine model of CLI, THNR administration can integrally re-establish blood perfusion with concomitant reduction of muscle damage and inflammation. Additionally, improvement of locomotor activities and motor coordination in ischemic limbs in THNR treated mice is also evident. Overall, the study demonstrates that THNR have the potential to be developed as an efficacious and cost-effective alternative clinical therapy for CLI, using a nanomedicine approach.
Collapse
Affiliation(s)
- Papia Basuthakur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Arpita Roy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Soumya Ghosh
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vincy Vijay
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debiprasad Sinha
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mydhili Radhakrishnan
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Arvind Kumar
- Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumana Chakravarty
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
18
|
Dong Y, Yang Y, Wang H, Feng D, Nist E, Yapundich N, Spurlock B, Craft M, Qian L, Liu J. Single-cell chromatin profiling reveals genetic programs activating proregenerative states in nonmyocyte cells. SCIENCE ADVANCES 2024; 10:eadk4694. [PMID: 38381829 PMCID: PMC10881044 DOI: 10.1126/sciadv.adk4694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 01/18/2024] [Indexed: 02/23/2024]
Abstract
Cardiac regeneration requires coordinated participation of multiple cell types whereby their communications result in transient activation of proregenerative cell states. Although the molecular characteristics and lineage origins of these activated cell states and their contribution to cardiac regeneration have been studied, the extracellular signaling and the intrinsic genetic program underlying the activation of the transient functional cell states remain largely unexplored. In this study, we delineated the chromatin landscapes of the noncardiomyocytes (nonCMs) of the regenerating heart at the single-cell level and inferred the cis-regulatory architectures and trans-acting factors that control cell type-specific gene expression programs. Moreover, further motif analysis and cell-specific genetic manipulations suggest that the macrophage-derived inflammatory signal tumor necrosis factor-α, acting via its downstream transcription factor complex activator protein-1, functions cooperatively with discrete transcription regulators to activate respective nonCM cell types critical for cardiac regeneration. Thus, our study defines the regulatory architectures and intercellular communication principles in zebrafish heart regeneration.
Collapse
Affiliation(s)
- Yanhan Dong
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yuchen Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Haofei Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Dong Feng
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Elizabeth Nist
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nicholas Yapundich
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian Spurlock
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Madison Craft
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
19
|
Wang P, Konja D, Singh S, Zhang B, Wang Y. Endothelial Senescence: From Macro- to Micro-Vasculature and Its Implications on Cardiovascular Health. Int J Mol Sci 2024; 25:1978. [PMID: 38396653 PMCID: PMC10889199 DOI: 10.3390/ijms25041978] [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: 12/27/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Endothelial cells line at the most inner layer of blood vessels. They act to control hemostasis, arterial tone/reactivity, wound healing, tissue oxygen, and nutrient supply. With age, endothelial cells become senescent, characterized by reduced regeneration capacity, inflammation, and abnormal secretory profile. Endothelial senescence represents one of the earliest features of arterial ageing and contributes to many age-related diseases. Compared to those in arteries and veins, endothelial cells of the microcirculation exhibit a greater extent of heterogeneity. Microcirculatory endothelial senescence leads to a declined capillary density, reduced angiogenic potentials, decreased blood flow, impaired barrier properties, and hypoperfusion in a tissue or organ-dependent manner. The heterogeneous phenotypes of microvascular endothelial cells in a particular vascular bed and across different tissues remain largely unknown. Accordingly, the mechanisms underlying macro- and micro-vascular endothelial senescence vary in different pathophysiological conditions, thus offering specific target(s) for therapeutic development of senolytic drugs.
Collapse
Affiliation(s)
- Peichun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sandeep Singh
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Beijia Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
20
|
Sunshine HL, Cicchetto AC, Kaczor-Urbanowicz KE, Ma F, Pi D, Symons C, Turner M, Shukla V, Christofk HR, Vallim TA, Iruela-Arispe ML. Endothelial Jagged1 levels and distribution are post-transcriptionally controlled by ZFP36 decay proteins. Cell Rep 2024; 43:113627. [PMID: 38157296 PMCID: PMC10884959 DOI: 10.1016/j.celrep.2023.113627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/02/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
Vascular morphogenesis requires a delicate gradient of Notch signaling controlled, in part, by the distribution of ligands (Dll4 and Jagged1). How Jagged1 (JAG1) expression is compartmentalized in the vascular plexus remains unclear. Here, we show that Jag1 mRNA is a direct target of zinc-finger protein 36 (ZFP36), an RNA-binding protein involved in mRNA decay that we find robustly induced by vascular endothelial growth factor (VEGF). Endothelial cells lacking ZFP36 display high levels of JAG1 and increase angiogenic sprouting in vitro. Furthermore, mice lacking Zfp36 in endothelial cells display mispatterned and increased levels of JAG1 in the developing retinal vascular plexus. Abnormal levels of JAG1 at the sprouting front alters NOTCH1 signaling, increasing the number of tip cells, a phenotype that is rescued by imposing haploinsufficiency of Jag1. Our findings reveal an important feedforward loop whereby VEGF stimulates ZFP36, consequently suppressing Jag1 to enable adequate levels of Notch signaling during sprouting angiogenesis.
Collapse
Affiliation(s)
- Hannah L Sunshine
- Molecular, Cellular, and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew C Cicchetto
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Karolina Elżbieta Kaczor-Urbanowicz
- Center for Oral and Head/Neck Oncology Research, UCLA Biosystems & Function, UCLA School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095-1668, USA; UCLA Section of Orthodontics, UCLA School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Feiyang Ma
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Danielle Pi
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Chloe Symons
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Martin Turner
- Immunology Programme, The Babraham Institute, CB22 3AT Cambridge, UK
| | - Vipul Shukla
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Center for Human Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Heather R Christofk
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095-1606, USA
| | - Thomas A Vallim
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095-1606, USA
| | - M Luisa Iruela-Arispe
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
21
|
Bhat GP, Maurizio A, Motta A, Podini P, Diprima S, Malpighi C, Brambilla I, Martins L, Badaloni A, Boselli D, Bianchi F, Pellegatta M, Genua M, Ostuni R, Del Carro U, Taveggia C, de Pretis S, Quattrini A, Bonanomi D. Structured wound angiogenesis instructs mesenchymal barrier compartments in the regenerating nerve. Neuron 2024; 112:209-229.e11. [PMID: 37972594 DOI: 10.1016/j.neuron.2023.10.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/19/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
Organ injury stimulates the formation of new capillaries to restore blood supply raising questions about the potential contribution of neoangiogenic vessel architecture to the healing process. Using single-cell mapping, we resolved the properties of endothelial cells that organize a polarized scaffold at the repair site of lesioned peripheral nerves. Transient reactivation of an embryonic guidance program is required to orient neovessels across the wound. Manipulation of this structured angiogenic response through genetic and pharmacological targeting of Plexin-D1/VEGF pathways within an early window of repair has long-term impact on configuration of the nerve stroma. Neovessels direct nerve-resident mesenchymal cells to mold a provisionary fibrotic scar by assembling an orderly system of stable barrier compartments that channel regenerating nerve fibers and shield them from the persistently leaky vasculature. Thus, guided and balanced repair angiogenesis enables the construction of a "bridge" microenvironment conducive for axon regrowth and homeostasis of the regenerated tissue.
Collapse
Affiliation(s)
- Ganesh Parameshwar Bhat
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Aurora Maurizio
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessia Motta
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Paola Podini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy; Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Santo Diprima
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy; Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Chiara Malpighi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Ilaria Brambilla
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Luis Martins
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Aurora Badaloni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Daniela Boselli
- FRACTAL-Flow cytometry Resource Advanced Cytometry Technical Applications Laboratory, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesca Bianchi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Marta Pellegatta
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Marco Genua
- San Raffaele Telethon Institute for Gene therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Renato Ostuni
- San Raffaele Telethon Institute for Gene therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ubaldo Del Carro
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Carla Taveggia
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Stefano de Pretis
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Angelo Quattrini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy; Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Dario Bonanomi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy.
| |
Collapse
|
22
|
Rehberger Likozar A, Ugovšek S, Šebeštjen M. Effects of proprotein convertase subtilisin-kexin type 9 inhibitors on inflammatory and hemostatic parameters in post myocardial infarction patients. Eur J Pharmacol 2024; 963:176232. [PMID: 38070635 DOI: 10.1016/j.ejphar.2023.176232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 01/07/2024]
Abstract
Despite progress in treatment, elevated levels of low-density lipoprotein cholesterol (LDL-C) and lipoprotein (a) (Lp(a)), represent a significant part of the residual risk. Both are associated with inflammation and the coagulation fibrinolytic system. The purpose of our research was to evaluate the effect of proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) on lipid parameters and indicators of inflammation, coagulation and fibrinolysis. We included 100 post myocardial infarction (MI) patients with insufficiently controlled LDL-C values despite the maximum dose of statin, and highly elevated Lp(a). Patients received alirocumab or evolocumab (150 mg sc or 140 mg sc every two weeks, respectively), or placebo for 6 months. In patients receiving PCSK9i, a significant decrease in total cholesterol (TC), LDL-C, triglycerides (TG) and Lp(a), and an increase in high density lipoprotein cholesterol (p < 0.001 for all) was found. Before treatment, the concentrations of TC, LDL-C and TG correlated with the concentrations of thrombin activatable fibrinolysis inhibitor (r = 0.41, p < 0.001; r = 0.353, p < 0.001; r = 0.311, p = 0.003, respectively), and plasminogen activator inhibitor-1 (r = 0.302, p = 0.007; r = 0.218, p = 0.049; r = 0.278; p = 0.013, respectively). The concentrations of TC and LDL-C correlated with overall fibrinolytic potential (r = -0.220, p = 0.034; r = -0.207, p = 0.047, respectively). The concentration of TG was related to the concentration of interleukin 6 (r = 0.290, p = 0.004) and interleukin 8 (r = 0.332, p = 0.001). No correlations between Lp(a) and inflammatory or hemostatic variables were found. No associations were found after treatment. Our results show that inflammatory cytokines and fibrinolytic parameters are related to LDL-C and not Lp(a) in post-MI patients before and with neither of them following PCSK9i treatment. The trial registration number: NCT04613167, Date of registration: November 3, 2020.
Collapse
Affiliation(s)
- Andreja Rehberger Likozar
- Department of Vascular Diseases, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| | - Sabina Ugovšek
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Department of Cardiology, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia
| | - Miran Šebeštjen
- Department of Vascular Diseases, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Department of Cardiology, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia.
| |
Collapse
|
23
|
Li Y, Liu Z, Han X, Liang F, Zhang Q, Huang X, Shi X, Huo H, Han M, Liu X, Zhu H, He L, Shen L, Hu X, Wang J, Wang QD, Smart N, Zhou B, He B. Dynamics of Endothelial Cell Generation and Turnover in Arteries During Homeostasis and Diseases. Circulation 2024; 149:135-154. [PMID: 38084582 DOI: 10.1161/circulationaha.123.064301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 10/06/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Endothelial cell (EC) generation and turnover by self-proliferation contributes to vascular repair and regeneration. The ability to accurately measure the dynamics of EC generation would advance our understanding of cellular mechanisms of vascular homeostasis and diseases. However, it is currently challenging to evaluate the dynamics of EC generation in large vessels such as arteries because of their infrequent proliferation. METHODS By using dual recombination systems based on Cre-loxP and Dre-rox, we developed a genetic system for temporally seamless recording of EC proliferation in vivo. We combined genetic recording of EC proliferation with single-cell RNA sequencing and gene knockout to uncover cellular and molecular mechanisms underlying EC generation in arteries during homeostasis and disease. RESULTS Genetic proliferation tracing reveals that ≈3% of aortic ECs undergo proliferation per month in adult mice during homeostasis. The orientation of aortic EC division is generally parallel to blood flow in the aorta, which is regulated by the mechanosensing protein Piezo1. Single-cell RNA sequencing analysis reveals 4 heterogeneous aortic EC subpopulations with distinct proliferative activity. EC cluster 1 exhibits transit-amplifying cell features with preferential proliferative capacity and enriched expression of stem cell markers such as Sca1 and Sox18. EC proliferation increases in hypertension but decreases in type 2 diabetes, coinciding with changes in the extent of EC cluster 1 proliferation. Combined gene knockout and proliferation tracing reveals that Hippo/vascular endothelial growth factor receptor 2 signaling pathways regulate EC proliferation in large vessels. CONCLUSIONS Genetic proliferation tracing quantitatively delineates the dynamics of EC generation and turnover, as well as EC division orientation, in large vessels during homeostasis and disease. An EC subpopulation in the aorta exhibits more robust cell proliferation during homeostasis and type 2 diabetes, identifying it as a potential therapeutic target for vascular repair and regeneration.
Collapse
Affiliation(s)
- Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Zixin Liu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Qianyu Zhang
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
| | - Xiuzhen Huang
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Xin Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Huanhuan Huo
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Maoying Han
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
| | - Xiuxiu Liu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Huan Zhu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China (L.H.)
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H., J.W.)
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H., J.W.)
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Q.D.W.)
| | - Nicola Smart
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, UK (N.S.)
| | - Bin Zhou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, China (B.Z.)
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| |
Collapse
|
24
|
Tanke NT, Liu Z, Gore MT, Bougaran P, Linares MB, Marvin A, Sharma A, Oatley M, Yu T, Quigley K, Vest S, Cook JG, Bautch VL. Endothelial cell flow-mediated quiescence is temporally regulated and utilizes the cell cycle inhibitor p27. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.09.544403. [PMID: 37662222 PMCID: PMC10473767 DOI: 10.1101/2023.06.09.544403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Background Endothelial cells regulate their cell cycle as blood vessels remodel and transition to quiescence downstream of blood flow-induced mechanotransduction. Laminar blood flow leads to quiescence, but how flow-mediated quiescence is established and maintained is poorly understood. Methods Primary human endothelial cells were exposed to laminar flow regimens and gene expression manipulations, and quiescence depth was analyzed via time to cell cycle re-entry after flow cessation. Mouse and zebrafish endothelial expression patterns were examined via scRNA seq analysis, and mutant or morphant fish lacking p27 were analyzed for endothelial cell cycle regulation and in vivo cellular behaviors. Results Arterial flow-exposed endothelial cells had a distinct transcriptome, and they first entered a deep quiescence, then transitioned to shallow quiescence under homeostatic maintenance conditions. In contrast, venous-flow exposed endothelial cells entered deep quiescence early that did not change with homeostasis. The cell cycle inhibitor p27 (CDKN1B) was required to establish endothelial flow-mediated quiescence, and expression levels positively correlated with quiescence depth. p27 loss in vivo led to endothelial cell cycle upregulation and ectopic sprouting, consistent with loss of quiescence. HES1 and ID3, transcriptional repressors of p27 upregulated by arterial flow, were required for quiescence depth changes and the reduced p27 levels associated with shallow quiescence. Conclusions Endothelial cell flow-mediated quiescence has unique properties and temporal regulation of quiescence depth that depends on the flow stimulus. These findings are consistent with a model whereby flow-mediated endothelial cell quiescence depth is temporally regulated downstream of p27 transcriptional regulation by HES1 and ID3. The findings are important in understanding endothelial cell quiescence mis-regulation that leads to vascular dysfunction and disease.
Collapse
Affiliation(s)
- Natalie T Tanke
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Ziqing Liu
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Michaelanthony T Gore
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Pauline Bougaran
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Mary B Linares
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Allison Marvin
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Arya Sharma
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Morgan Oatley
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Tianji Yu
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Kaitlyn Quigley
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Sarah Vest
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Jeanette Gowen Cook
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Victoria L Bautch
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
25
|
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
|
26
|
Konishi H, Rahmawati FN, Okamoto N, Akuta K, Inukai K, Jia W, Muramatsu F, Takakura N. Discovery of Transcription Factors Involved in the Maintenance of Resident Vascular Endothelial Stem Cell Properties. Mol Cell Biol 2024; 44:17-26. [PMID: 38247234 PMCID: PMC10829836 DOI: 10.1080/10985549.2023.2297997] [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: 06/09/2023] [Accepted: 12/08/2023] [Indexed: 01/23/2024] Open
Abstract
A resident vascular endothelial stem cell (VESC) population expressing CD157 has been identified recently in mice. Herein, we identified transcription factors (TFs) regulating CD157 expression in endothelial cells (ECs) that were associated with drug resistance, angiogenesis, and EC proliferation. In the first screening, we detected 20 candidate TFs through the CD157 promoter and gene expression analyses. We found that 10 of the 20 TFs induced CD157 expression in ECs. We previously reported that 70% of CD157 VESCs were side population (SP) ECs that abundantly expressed ATP-binding cassette (ABC) transporters. Here, we found that the 10 TFs increased the expression of several ABC transporters in ECs and increased the proportion of SP ECs. Of these 10 TFs, we found that six (Atf3, Bhlhe40, Egr1, Egr2, Elf3, and Klf4) were involved in the manifestation of the SP phenotype. Furthermore, the six TFs enhanced tube formation and proliferation in ECs. Single-cell RNA sequence data in liver ECs suggested that Atf3 and Klf4 contributed to the production of CD157+ VESCs in the postnatal period. We concluded that Klf4 might be important for the development and maintenance of liver VESCs. Our work suggests that a TF network is involved in the differentiation hierarchy of VESCs.
Collapse
Affiliation(s)
- Hirotaka Konishi
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Fitriana N. Rahmawati
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Naoki Okamoto
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Keigo Akuta
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Koichi Inukai
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Weizhen Jia
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Fumitaka Muramatsu
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Signal Transduction, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Japan
- Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| |
Collapse
|
27
|
Molnár AÁ, Pásztor DT, Tarcza Z, Merkely B. Cells in Atherosclerosis: Focus on Cellular Senescence from Basic Science to Clinical Practice. Int J Mol Sci 2023; 24:17129. [PMID: 38138958 PMCID: PMC10743093 DOI: 10.3390/ijms242417129] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Aging is a major risk factor of atherosclerosis through different complex pathways including replicative cellular senescence and age-related clonal hematopoiesis. In addition to aging, extracellular stress factors, such as mechanical and oxidative stress, can induce cellular senescence, defined as premature cellular senescence. Senescent cells can accumulate within atherosclerotic plaques over time and contribute to plaque instability. This review summarizes the role of cellular senescence in the complex pathophysiology of atherosclerosis and highlights the most important senotherapeutics tested in cardiovascular studies targeting senescence. Continued bench-to-bedside research in cellular senescence might allow the future implementation of new effective anti-atherosclerotic preventive and treatment strategies in clinical practice.
Collapse
Affiliation(s)
- Andrea Ágnes Molnár
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary; (D.T.P.); (Z.T.); (B.M.)
| | | | | | | |
Collapse
|
28
|
Mahara A, Shirai M, Soni R, Le HT, Shimizu K, Hirano Y, Yamaoka T. Vascular tissue reconstruction by monocyte subpopulations on small-diameter acellular grafts via integrin activation. Mater Today Bio 2023; 23:100847. [PMID: 37953756 PMCID: PMC10632538 DOI: 10.1016/j.mtbio.2023.100847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023] Open
Abstract
Although the clinical application of cell-free tissue-engineered vascular grafts (TEVGs) has been proposed, vascular tissue regeneration mechanisms have not been fully clarified. Here, we report that monocyte subpopulations reconstruct vascular-like tissues through integrin signaling. An Arg-Glu-Asp-Val peptide-modified acellular long-bypass graft was used as the TEVG, and tissue regeneration in the graft was evaluated using a cardiopulmonary pump system and porcine transplantation model. In 1 day, the luminal surface of the graft was covered with cells that expressed CD163, CD14, and CD16, which represented the monocyte subpopulation, and they exhibited proliferative and migratory abilities. RNA sequencing showed that captured cells had an immune-related phenotype similar to that of monocytes and strongly expressed cell adhesion-related genes. In vitro angiogenesis assay showed that tube formation of the captured cells occurred via integrin signal activation. After medium- and long-term graft transplantation, the captured cells infiltrated the tunica media layer and constructed vascular with a CD31/CD105-positive layer and an αSMA-positive structure after 3 months. This finding, including multiple early-time observations provides clear evidence that blood-circulating monocytes are directly involved in vascular remodeling.
Collapse
Affiliation(s)
- Atsushi Mahara
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Manabu Shirai
- Omics Research Center, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Raghav Soni
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Hue Thi Le
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Kaito Shimizu
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamatecho, Suita, Osaka, 565-8680, Japan
| | - Yoshiaki Hirano
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamatecho, Suita, Osaka, 565-8680, Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| |
Collapse
|
29
|
Hu J, Leisegang MS, Looso M, Drekolia MK, Wittig J, Mettner J, Karantanou C, Kyselova A, Dumbovic G, Li X, Li Y, Guenther S, John D, Siragusa M, Zukunft S, Oo JA, Wittig I, Hille S, Weigert A, Knapp S, Brandes RP, Müller OJ, Papapetropoulos A, Sigala F, Dobreva G, Kojonazarov B, Fleming I, Bibli SI. Disrupted Binding of Cystathionine γ-Lyase to p53 Promotes Endothelial Senescence. Circ Res 2023; 133:842-857. [PMID: 37800327 DOI: 10.1161/circresaha.123.323084] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Advanced age is unequivocally linked to the development of cardiovascular disease; however, the mechanisms resulting in reduced endothelial cell regeneration remain poorly understood. Here, we investigated novel mechanisms involved in endothelial cell senescence that impact endothelial cell transcription and vascular repair after injury. METHODS Native endothelial cells were isolated from young (20±3.4 years) and aged (80±2.3 years) individuals and subjected to molecular analyses to assess global transcriptional and metabolic changes. In vitro studies were conducted using primary human and murine endothelial cells. A murine aortic re-endothelialization model was used to examine endothelial cell regenerative capacity in vivo. RESULTS RNA sequencing of native endothelial cells revealed that aging resulted in p53-mediated reprogramming to express senescence-associated genes and suppress glycolysis. Reduced glucose uptake and ATP contributed to attenuated assembly of the telomerase complex, which was required for endothelial cell proliferation. Enhanced p53 activity in aging was linked to its acetylation on K120 due to enhanced activity of the acetyltransferase MOZ (monocytic leukemic zinc finger). Mechanistically, p53 acetylation and translocation were, at least partially, attributed to the loss of the vasoprotective enzyme, CSE (cystathionine γ-lyase). CSE physically anchored p53 in the cytosol to prevent its nuclear translocation and CSE absence inhibited AKT (Protein kinase B)-mediated MOZ phosphorylation, which in turn increased MOZ activity and subsequently p53 acetylation. In mice, the endothelial cell-specific deletion of CSE activated p53, induced premature endothelial senescence, and arrested vascular repair after injury. In contrast, the adeno-associated virus 9-mediated re-expression of an active CSE mutant retained p53 in the cytosol, maintained endothelial glucose metabolism and proliferation, and prevented endothelial cell senescence. Adenoviral overexpression of CSE in native endothelial cells from aged individuals maintained low p53 activity and reactivated telomerase to revert endothelial cell senescence. CONCLUSIONS Aging-associated impairment of vascular repair is partly determined by the vasoprotective enzyme CSE.
Collapse
Affiliation(s)
- Jiong Hu
- Department of Histology and Embryology, School of Basic Medicine (J.H., X.L., Y.L.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Sino-German Laboratory of CardioPulmonary Science (J.H., I.F.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Matthias S Leisegang
- Institute for Cardiovascular Physiology (M.S.L., J.A.O., R.P.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Mario Looso
- Bioinformatics Core Unit, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.L., S.G.)
- German Center for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt am Main (M.L., S.G., R.P.B., I.F., S.-I.B.)
| | - Maria-Kyriaki Drekolia
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Janina Wittig
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Janina Mettner
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Christina Karantanou
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Anastasia Kyselova
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Gabrjela Dumbovic
- Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (G.D.)
| | - Xiaoming Li
- Department of Histology and Embryology, School of Basic Medicine (J.H., X.L., Y.L.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Yuanyuan Li
- Department of Histology and Embryology, School of Basic Medicine (J.H., X.L., Y.L.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Stefan Guenther
- Bioinformatics Core Unit, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.L., S.G.)
- German Center for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt am Main (M.L., S.G., R.P.B., I.F., S.-I.B.)
| | - David John
- Institute of Cardiovascular Regeneration (D.J.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Sven Zukunft
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - James A Oo
- Institute for Cardiovascular Physiology (M.S.L., J.A.O., R.P.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ilka Wittig
- Sino-German Laboratory of CardioPulmonary Science (J.H., I.F.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Functional Proteomics, Institute for Cardiovascular Physiology (I.W.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Susanne Hille
- Department of Internal Medicine III, University of Kiel, Germany (S.H., O.J.M.)
| | - Andreas Weigert
- Institute of Biochemistry I (A.W.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry and Buchmann Institute for Molecular Life Sciences (S.K.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology (M.S.L., J.A.O., R.P.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt am Main (M.L., S.G., R.P.B., I.F., S.-I.B.)
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, Germany (S.H., O.J.M.)
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany (O.J.M.)
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy (A.P.), National and Kapodistrian University of Athens, Greece
| | - Fragiska Sigala
- First Propedeutic Department of Surgery, Vascular Surgery Division (F.S.), National and Kapodistrian University of Athens, Greece
| | - Gergana Dobreva
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (G.D.)
| | - Baktybek Kojonazarov
- Institute for Lung Health (ILH) (B.K.), Justus Liebig University, Giessen, Germany
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI) (B.K.), Justus Liebig University, Giessen, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Sofia-Iris Bibli
- Institute for Vascular Signalling, Centre for Molecular Medicine (J.H., M.-K.D., J.W., J.M., C.K., A.K., X.L., M.S., S.Z., I.F., S.-I.B.), Goethe University Frankfurt, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt am Main (M.L., S.G., R.P.B., I.F., S.-I.B.)
| |
Collapse
|
30
|
Pohl L, Schiessl IM. Endothelial cell plasticity in kidney fibrosis and disease. Acta Physiol (Oxf) 2023; 239:e14038. [PMID: 37661749 DOI: 10.1111/apha.14038] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/29/2023] [Accepted: 08/11/2023] [Indexed: 09/05/2023]
Abstract
Renal endothelial cells demonstrate an impressive remodeling potential during angiogenic sprouting, vessel repair or while transitioning into mesenchymal cells. These different processes may play important roles in both renal disease progression or regeneration while underlying signaling pathways of different endothelial cell plasticity routes partly overlap. Angiogenesis contributes to wound healing after kidney injury and pharmaceutical modulation of angiogenesis may home a great therapeutic potential. Yet, it is not clear whether any differentiated endothelial cell can proliferate or whether regenerative processes are largely controlled by resident or circulating endothelial progenitor cells. In the glomerular compartment for example, a distinct endothelial progenitor cell population may remodel the glomerular endothelium after injury. Endothelial-to-mesenchymal transition (EndoMT) in the kidney is vastly documented and often associated with endothelial dysfunction, fibrosis, and kidney disease progression. Especially the role of EndoMT in renal fibrosis is controversial. Studies on EndoMT in vivo determined possible conclusions on the pathophysiological role of EndoMT in the kidney, but whether endothelial cells really contribute to kidney fibrosis and if not what other cellular and functional outcomes derive from EndoMT in kidney disease is unclear. Sequencing data, however, suggest no participation of endothelial cells in extracellular matrix deposition. Thus, more in-depth classification of cellular markers and the fate of EndoMT cells in the kidney is needed. In this review, we describe different signaling pathways of endothelial plasticity, outline methodological approaches and evidence for functional and structural implications of angiogenesis and EndoMT in the kidney, and eventually discuss controversial aspects in the literature.
Collapse
Affiliation(s)
- Layla Pohl
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
31
|
Dudley AC, Griffioen AW. Pathological angiogenesis: mechanisms and therapeutic strategies. Angiogenesis 2023; 26:313-347. [PMID: 37060495 PMCID: PMC10105163 DOI: 10.1007/s10456-023-09876-7] [Citation(s) in RCA: 185] [Impact Index Per Article: 92.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/26/2023] [Indexed: 04/16/2023]
Abstract
In multicellular organisms, angiogenesis, the formation of new blood vessels from pre-existing ones, is an essential process for growth and development. Different mechanisms such as vasculogenesis, sprouting, intussusceptive, and coalescent angiogenesis, as well as vessel co-option, vasculogenic mimicry and lymphangiogenesis, underlie the formation of new vasculature. In many pathological conditions, such as cancer, atherosclerosis, arthritis, psoriasis, endometriosis, obesity and SARS-CoV-2(COVID-19), developmental angiogenic processes are recapitulated, but are often done so without the normal feedback mechanisms that regulate the ordinary spatial and temporal patterns of blood vessel formation. Thus, pathological angiogenesis presents new challenges yet new opportunities for the design of vascular-directed therapies. Here, we provide an overview of recent insights into blood vessel development and highlight novel therapeutic strategies that promote or inhibit the process of angiogenesis to stabilize, reverse, or even halt disease progression. In our review, we will also explore several additional aspects (the angiogenic switch, hypoxia, angiocrine signals, endothelial plasticity, vessel normalization, and endothelial cell anergy) that operate in parallel to canonical angiogenesis mechanisms and speculate how these processes may also be targeted with anti-angiogenic or vascular-directed therapies.
Collapse
Affiliation(s)
- Andrew C Dudley
- Department of Microbiology, Immunology and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA.
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
32
|
Pleskova SN, Bezrukov NA, Gorshkova EN, Bobyk SZ, Lazarenko EV. Exploring the Process of Neutrophil Transendothelial Migration Using Scanning Ion-Conductance Microscopy. Cells 2023; 12:1806. [PMID: 37443839 PMCID: PMC10340179 DOI: 10.3390/cells12131806] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023] Open
Abstract
The dynamics of neutrophil transendothelial migration was investigated in a model of experimental septicopyemia. Scanning ion-conductance microscopy allowed us to determine changes in morphometric characteristics of endothelial cells during this process. In the presence of a pyogenic lesion simulated by Staphylococcus aureus, such migration was accompanied by both compensatory reactions and alteration of both neutrophils and endothelial cells. Neutrophils demonstrated crawling along the contact sites between endothelial cells, swarming phenomenon, as well as anergy and formation of neutrophil extracellular traps (NETs) as a normergic state. Neutrophil swarming was accompanied by an increase in the intercellular spaces between endothelial cells. Endothelial cells decreased the area of adhesion to the substrate, which was determined by a decrease in the cell projection area, and the cell membrane was smoothed. However, endothelial cell rigidity was paradoxically unchanged compared to the control. Over time, neutrophil migration led to a more significant alteration of endothelial cells: first, shallow perforations in the membrane were formed, which were repaired rather quickly, then stress fibrils were formed, and finally, endothelial cells died and multiple perforations were formed on their membrane.
Collapse
Affiliation(s)
- Svetlana N. Pleskova
- Research Laboratory of Scanning Probe Microscopy, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (N.A.B.); (E.N.G.); (S.Z.B.); (E.V.L.)
- Department “Nanotechnology and Biotechnology”, Nizhny Novgorod State Technical University n. a. R.E. Alekseev, 603115 Nizhny Novgorod, Russia
| | - Nikolay A. Bezrukov
- Research Laboratory of Scanning Probe Microscopy, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (N.A.B.); (E.N.G.); (S.Z.B.); (E.V.L.)
| | - Ekaterina N. Gorshkova
- Research Laboratory of Scanning Probe Microscopy, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (N.A.B.); (E.N.G.); (S.Z.B.); (E.V.L.)
| | - Sergey Z. Bobyk
- Research Laboratory of Scanning Probe Microscopy, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (N.A.B.); (E.N.G.); (S.Z.B.); (E.V.L.)
| | - Ekaterina V. Lazarenko
- Research Laboratory of Scanning Probe Microscopy, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (N.A.B.); (E.N.G.); (S.Z.B.); (E.V.L.)
| |
Collapse
|
33
|
Rodor J, Chen SH, Baker AH. Response to: Are endothelial cell proliferation and mesenchymal transition as distinguishing characteristics of 3-week Sugen5416/hypoxia mice model? Cardiovasc Res 2023; 119:e142-e143. [PMID: 37170759 PMCID: PMC10325695 DOI: 10.1093/cvr/cvad075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/13/2023] Open
Affiliation(s)
- Julie Rodor
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Shiau-Haln Chen
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Andrew H Baker
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
34
|
Thompson M, Sakabe M, Verba M, Hao J, Meadows SM, Lu QR, Xin M. PRDM16 regulates arterial development and vascular integrity. Front Physiol 2023; 14:1165379. [PMID: 37324380 PMCID: PMC10267475 DOI: 10.3389/fphys.2023.1165379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023] Open
Abstract
Proper vascular formation is regulated by multiple signaling pathways. The vascular endothelial growth factor (VEGF) signaling promotes endothelial proliferation. Notch and its downstream targets act to lead endothelial cells toward an arterial fate through regulation of arterial gene expression. However, the mechanisms of how endothelial cells (ECs) in the artery maintain their arterial characteristics remain unclear. Here, we show that PRDM16 (positive regulatory domain-containing protein 16), a zinc finger transcription factor, is expressed in arterial ECs, but not venous ECs in developing embryos and neonatal retinas. Endothelial-specific deletion of Prdm16 induced ectopic venous marker expression in the arterial ECs and reduced vascular smooth muscle cell (vSMC) recruitment around arteries. Whole-genome transcriptome analysis using isolated brain ECs show that the expression of Angpt2 (encoding ANGIOPOIETIN2, which inhibits vSMC recruitment) is upregulated in the Prdm16 knockout ECs. Conversely, forced expression of PRDM16 in venous ECs is sufficient to induce arterial gene expression and repress the ANGPT2 level. Together, these results reveal an arterial cell-autonomous function for PRDM16 in suppressing venous characteristics in arterial ECs.
Collapse
Affiliation(s)
- Michael Thompson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Masahide Sakabe
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Mark Verba
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Jiukuan Hao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Stryder M. Meadows
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA, United States
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States
| | - Q. Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Mei Xin
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
35
|
Cicchetto AC, Jacobson EC, Sunshine H, Wilde BR, Krall AS, Jarrett KE, Sedgeman L, Turner M, Plath K, Iruela-Arispe ML, de Aguiar Vallim TQ, Christofk HR. ZFP36-mediated mRNA decay regulates metabolism. Cell Rep 2023; 42:112411. [PMID: 37086408 PMCID: PMC10332406 DOI: 10.1016/j.celrep.2023.112411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/17/2023] [Accepted: 04/04/2023] [Indexed: 04/23/2023] Open
Abstract
Cellular metabolism is tightly regulated by growth factor signaling, which promotes metabolic rewiring to support growth and proliferation. While growth factor-induced transcriptional and post-translational modes of metabolic regulation have been well defined, whether post-transcriptional mechanisms impacting mRNA stability regulate this process is less clear. Here, we present the ZFP36/L1/L2 family of RNA-binding proteins and mRNA decay factors as key drivers of metabolic regulation downstream of acute growth factor signaling. We quantitatively catalog metabolic enzyme and nutrient transporter mRNAs directly bound by ZFP36 following growth factor stimulation-many of which encode rate-limiting steps in metabolic pathways. Further, we show that ZFP36 directly promotes the mRNA decay of Enolase 2 (Eno2), altering Eno2 protein expression and enzymatic activity, and provide evidence of a ZFP36/Eno2 axis during VEGF-stimulated developmental retinal angiogenesis. Thus, ZFP36-mediated mRNA decay serves as an important mode of metabolic regulation downstream of growth factor signaling within dynamic cell and tissue states.
Collapse
Affiliation(s)
- Andrew C Cicchetto
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Elsie C Jacobson
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Hannah Sunshine
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Blake R Wilde
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Abigail S Krall
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Kelsey E Jarrett
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Department of Medicine, Division of Cardiology, UCLA, Los Angeles, CA, USA
| | - Leslie Sedgeman
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Department of Medicine, Division of Cardiology, UCLA, Los Angeles, CA, USA
| | - Martin Turner
- Immunology Programme, The Babraham Institute, Cambridge, UK
| | - Kathrin Plath
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - M Luisa Iruela-Arispe
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Thomas Q de Aguiar Vallim
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Department of Medicine, Division of Cardiology, UCLA, Los Angeles, CA, USA; Molecular Biology Institute, UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Heather R Christofk
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
36
|
Niethamer TK, Levin LI, Morley MP, Babu A, Zhou S, Morrisey EE. Atf3 defines a population of pulmonary endothelial cells essential for lung regeneration. eLife 2023; 12:e83835. [PMID: 37233732 PMCID: PMC10219650 DOI: 10.7554/elife.83835] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
Following acute injury, the capillary vascular bed in the lung must be repaired to reestablish gas exchange with the external environment. Little is known about the transcriptional and signaling factors that drive pulmonary endothelial cell (EC) proliferation and subsequent regeneration of pulmonary capillaries, as well as their response to stress. Here, we show that the transcription factor Atf3 is essential for the regenerative response of the mouse pulmonary endothelium after influenza infection. Atf3 expression defines a subpopulation of capillary ECs enriched in genes involved in endothelial development, differentiation, and migration. During lung alveolar regeneration, this EC population expands and increases the expression of genes involved in angiogenesis, blood vessel development, and cellular response to stress. Importantly, endothelial cell-specific loss of Atf3 results in defective alveolar regeneration, in part through increased apoptosis and decreased proliferation in the endothelium. This leads to the general loss of alveolar endothelium and persistent morphological changes to the alveolar niche, including an emphysema-like phenotype with enlarged alveolar airspaces lined with regions that lack vascular investment. Taken together, these data implicate Atf3 as an essential component of the vascular response to acute lung injury that is required for successful lung alveolar regeneration.
Collapse
Affiliation(s)
- Terren K Niethamer
- Department of MedicinePhiladelphiaUnited States
- Department of Cell and Developmental BiologyPhiladelphiaUnited States
- Penn-Children’s Hospital of Philadelphia Lung Biology Institute, University of PennsylvaniaPhiladelphiaUnited States
- Penn Cardiovascular Institute, University of PennsylvaniaPhiladelphiaUnited States
| | - Lillian I Levin
- Department of MedicinePhiladelphiaUnited States
- Penn Cardiovascular Institute, University of PennsylvaniaPhiladelphiaUnited States
| | - Michael P Morley
- Department of MedicinePhiladelphiaUnited States
- Penn-Children’s Hospital of Philadelphia Lung Biology Institute, University of PennsylvaniaPhiladelphiaUnited States
- Penn Cardiovascular Institute, University of PennsylvaniaPhiladelphiaUnited States
| | - Apoorva Babu
- Department of MedicinePhiladelphiaUnited States
- Penn-Children’s Hospital of Philadelphia Lung Biology Institute, University of PennsylvaniaPhiladelphiaUnited States
- Penn Cardiovascular Institute, University of PennsylvaniaPhiladelphiaUnited States
| | - Su Zhou
- Department of MedicinePhiladelphiaUnited States
- Penn Cardiovascular Institute, University of PennsylvaniaPhiladelphiaUnited States
| | - Edward E Morrisey
- Department of MedicinePhiladelphiaUnited States
- Department of Cell and Developmental BiologyPhiladelphiaUnited States
- Penn-Children’s Hospital of Philadelphia Lung Biology Institute, University of PennsylvaniaPhiladelphiaUnited States
- Penn Cardiovascular Institute, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
37
|
Sum H, Brewer AC. Epigenetic modifications as therapeutic targets in atherosclerosis: a focus on DNA methylation and non-coding RNAs. Front Cardiovasc Med 2023; 10:1183181. [PMID: 37304954 PMCID: PMC10248074 DOI: 10.3389/fcvm.2023.1183181] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/02/2023] [Indexed: 06/13/2023] Open
Abstract
Significant progress in the diagnosis and treatment of cardiovascular disease (CVD) has been made in the past decade, yet it remains a leading cause of morbidity and mortality globally, claiming an estimated 17.9 million deaths per year. Although encompassing any condition that affects the circulatory system, including thrombotic blockage, stenosis, aneurysms, blood clots and arteriosclerosis (general hardening of the arteries), the most prevalent underlying hallmark of CVD is atherosclerosis; the plaque-associated arterial thickening. Further, distinct CVD conditions have overlapping dysregulated molecular and cellular characteristics which underlie their development and progression, suggesting some common aetiology. The identification of heritable genetic mutations associated with the development of atherosclerotic vascular disease (AVD), in particular resulting from Genome Wide Association Studies (GWAS) studies has significantly improved the ability to identify individuals at risk. However, it is increasingly recognised that environmentally-acquired, epigenetic changes are key factors associated with atherosclerosis development. Increasing evidence suggests that these epigenetic changes, most notably DNA methylation and the misexpression of non-coding, microRNAs (miRNAs) are potentially both predictive and causal in AVD development. This, together with their reversible nature, makes them both useful biomarkers for disease and attractive therapeutic targets potentially to reverse AVD progression. We consider here the association of aberrant DNA methylation and dysregulated miRNA expression with the aetiology and progression of atherosclerosis, and the potential development of novel cell-based strategies to target these epigenetic changes therapeutically.
Collapse
|
38
|
Kam CY, Singh ID, Gonzalez DG, Matte-Martone C, Solá P, Solanas G, Bonjoch J, Marsh E, Hirschi KK, Greco V. Mechanisms of skin vascular maturation and maintenance captured by longitudinal imaging of live mice. Cell 2023; 186:2345-2360.e16. [PMID: 37167971 PMCID: PMC10225355 DOI: 10.1016/j.cell.2023.04.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/03/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023]
Abstract
A functional network of blood vessels is essential for organ growth and homeostasis, yet how the vasculature matures and maintains homeostasis remains elusive in live mice. By longitudinally tracking the same neonatal endothelial cells (ECs) over days to weeks, we found that capillary plexus expansion is driven by vessel regression to optimize network perfusion. Neonatal ECs rearrange positions to evenly distribute throughout the developing plexus and become positionally stable in adulthood. Upon local ablation, adult ECs survive through a plasmalemmal self-repair response, while neonatal ECs are predisposed to die. Furthermore, adult ECs reactivate migration to assist vessel repair. Global ablation reveals coordinated maintenance of the adult vascular architecture that allows for eventual network recovery. Lastly, neonatal remodeling and adult maintenance of the skin vascular plexus are orchestrated by temporally restricted, neonatal VEGFR2 signaling. Our work sheds light on fundamental mechanisms that underlie both vascular maturation and adult homeostasis in vivo.
Collapse
Affiliation(s)
- Chen Yuan Kam
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ishani D Singh
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - David G Gonzalez
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Paloma Solá
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Guiomar Solanas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Júlia Bonjoch
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Edward Marsh
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Valentina Greco
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Cell Biology and Dermatology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
39
|
Parab S, Setten E, Astanina E, Bussolino F, Doronzo G. The tissue-specific transcriptional landscape underlines the involvement of endothelial cells in health and disease. Pharmacol Ther 2023; 246:108418. [PMID: 37088448 DOI: 10.1016/j.pharmthera.2023.108418] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 03/23/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Endothelial cells (ECs) that line vascular and lymphatic vessels are being increasingly recognized as important to organ function in health and disease. ECs participate not only in the trafficking of gases, metabolites, and cells between the bloodstream and tissues but also in the angiocrine-based induction of heterogeneous parenchymal cells, which are unique to their specific tissue functions. The molecular mechanisms regulating EC heterogeneity between and within different tissues are modeled during embryogenesis and become fully established in adults. Any changes in adult tissue homeostasis induced by aging, stress conditions, and various noxae may reshape EC heterogeneity and induce specific transcriptional features that condition a functional phenotype. Heterogeneity is sustained via specific genetic programs organized through the combinatory effects of a discrete number of transcription factors (TFs) that, at the single tissue-level, constitute dynamic networks that are post-transcriptionally and epigenetically regulated. This review is focused on outlining the TF-based networks involved in EC specialization and physiological and pathological stressors thought to modify their architecture.
Collapse
Affiliation(s)
- Sushant Parab
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Elisa Setten
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Elena Astanina
- Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy.
| | - Gabriella Doronzo
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| |
Collapse
|
40
|
Kotlyarov S. Effects of Atherogenic Factors on Endothelial Cells: Bioinformatics Analysis of Differentially Expressed Genes and Signaling Pathways. Biomedicines 2023; 11:1216. [PMID: 37189834 PMCID: PMC10135807 DOI: 10.3390/biomedicines11041216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
(1) Background: Atherosclerosis is a serious medical condition associated with high morbidity and mortality rates. It develops over many years as a complex chain of events in the vascular wall involving various cells and is influenced by many factors of clinical interest. (2) Methods: In this study, we performed a bioinformatic analysis of Gene Expression Omnibus (GEO) datasets to investigate the gene ontology of differentially expressed genes (DEGs) in endothelial cells exposed to atherogenic factors such as tobacco smoking, oscillatory shear, and oxidized low-density lipoproteins (oxLDL). DEGs were identified using the limma R package, and gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and protein-protein interaction (PPI) network analysis were performed. (3) Results: We studied biological processes and signaling pathways involving DEGs in endothelial cells under the influence of atherogenic factors. GO enrichment analysis demonstrated that the DEGs were mainly involved in cytokine-mediated signaling pathway, innate immune response, lipid biosynthetic process, 5-lipoxygenase activity, and nitric-oxide synthase activity. KEGG pathway enrichment analysis showed that common pathways included tumor necrosis factor signaling pathway, NF-κB signaling pathway, NOD-like receptor signaling pathway, lipid and atherosclerosis, lipoprotein particle binding, and apoptosis. (4) Conclusions: Atherogenic factors such as smoking, impaired flow, and oxLDL contribute to impaired innate immune response, metabolism, and apoptosis in endothelial cells, potentially leading to the development of atherosclerosis.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
41
|
Marzoog BA. Endothelial cell autophagy in the context of disease development. Anat Cell Biol 2023; 56:16-24. [PMID: 36267005 PMCID: PMC9989784 DOI: 10.5115/acb.22.098] [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: 05/11/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 11/27/2022] Open
Abstract
Endothelial cells (EC) are the anatomical boundaries between the intravascular and extravascular space. Damage to ECs is catastrophic and induces endothelial cell dysfunction. The pathogenesis is multifactorial and involves dysregulation in the signaling pathways, membrane lipids ratio disturbance, cell-cell adhesion disturbance, unfolded protein response, lysosomal and mitochondrial stress, autophagy dysregulation, and oxidative stress. Autophagy is a lysosomal-dependent turnover of intracellular components. Autophagy was recognized early in the pathogenesis of endothelial dysfunction. Autophagy is a remarkable patho (physiological) process in the cell homeostasis regulation including EC. Regulation of autophagy rate is disease-dependent and impaired with aging. Up-regulation of autophagy induces endothelial cell regeneration/differentiation and improves the function of impaired ones. The paper scrutinizes the molecular mechanisms and triggers of EC dysregulation and current perspectives for future therapeutic strategies by autophagy targeting.
Collapse
|
42
|
Becker LM, Chen SH, Rodor J, de Rooij LPMH, Baker AH, Carmeliet P. Deciphering endothelial heterogeneity in health and disease at single-cell resolution: progress and perspectives. Cardiovasc Res 2023; 119:6-27. [PMID: 35179567 PMCID: PMC10022871 DOI: 10.1093/cvr/cvac018] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/16/2021] [Accepted: 02/16/2022] [Indexed: 11/14/2022] Open
Abstract
Endothelial cells (ECs) constitute the inner lining of vascular beds in mammals and are crucial for homeostatic regulation of blood vessel physiology, but also play a key role in pathogenesis of many diseases, thereby representing realistic therapeutic targets. However, it has become evident that ECs are heterogeneous, encompassing several subtypes with distinct functions, which makes EC targeting and modulation in diseases challenging. The rise of the new single-cell era has led to an emergence of studies aimed at interrogating transcriptome diversity along the vascular tree, and has revolutionized our understanding of EC heterogeneity from both a physiological and pathophysiological context. Here, we discuss recent landmark studies aimed at teasing apart the heterogeneous nature of ECs. We cover driving (epi)genetic, transcriptomic, and metabolic forces underlying EC heterogeneity in health and disease, as well as current strategies used to combat disease-enriched EC phenotypes, and propose strategies to transcend largely descriptive heterogeneity towards prioritization and functional validation of therapeutically targetable drivers of EC diversity. Lastly, we provide an overview of the most recent advances and hurdles in single EC OMICs.
Collapse
Affiliation(s)
| | | | | | | | - Andrew H Baker
- Corresponding authors. Tel: +32 16 32 62 47, E-mail: (P.C.); Tel: +44 (0)131 242 6774, E-mail: (A.H.B.)
| | - Peter Carmeliet
- Corresponding authors. Tel: +32 16 32 62 47, E-mail: (P.C.); Tel: +44 (0)131 242 6774, E-mail: (A.H.B.)
| |
Collapse
|
43
|
Lin Y, Banno K, Gil CH, Myslinski J, Hato T, Shelley WC, Gao H, Xuei X, Liu Y, Basile DP, Yoshimoto M, Prasain N, Tarnawsky SP, Adams RH, Naruse K, Yoshida J, Murphy MP, Horie K, Yoder MC. Origin, prospective identification, and function of circulating endothelial colony-forming cells in mice and humans. JCI Insight 2023; 8:e164781. [PMID: 36692963 PMCID: PMC10077473 DOI: 10.1172/jci.insight.164781] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Most circulating endothelial cells are apoptotic, but rare circulating endothelial colony-forming cells (C-ECFCs), also known as blood outgrowth endothelial cells, with proliferative and vasculogenic activity can be cultured; however, the origin and naive function of these C-ECFCs remains obscure. Herein, detailed lineage tracing revealed murine C-ECFCs emerged in the early postnatal period, displayed high vasculogenic potential with enriched frequency of clonal proliferative cells compared with tissue-resident ECFCs, and were not committed to or derived from the BM hematopoietic system but from tissue-resident ECFCs. In humans, C-ECFCs were present in the CD34bright cord blood mononuclear subset, possessed proliferative potential and in vivo vasculogenic function in a naive or cultured state, and displayed a single cell transcriptome sharing some umbilical venous endothelial cell features, such as a higher protein C receptor and extracellular matrix gene expression. This study provides an advance for the field by identifying the origin, naive function, and antigens to prospectively isolate C-ECFCs for translational studies.
Collapse
Affiliation(s)
- Yang Lin
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Kimihiko Banno
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Physiology II, Nara Medical University, Kashihara, Nara, Japan
| | - Chang-Hyun Gil
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery
| | | | | | - William C. Shelley
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, and
| | | | - Yunlong Liu
- Department of Medical and Molecular Genetics, and
| | - David P. Basile
- Department of Anatomy Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Momoko Yoshimoto
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nutan Prasain
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Stefan P. Tarnawsky
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ralf H. Adams
- Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Katsuhiko Naruse
- Department of Obstetrics & Gynecology, Nara Medical University, Kashihara, Nara, Japan
| | - Junko Yoshida
- Department of Physiology II, Nara Medical University, Kashihara, Nara, Japan
| | | | - Kyoji Horie
- Department of Physiology II, Nara Medical University, Kashihara, Nara, Japan
| | - Mervin C. Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
44
|
Abstract
Vascular endothelial cells form the inner layer of blood vessels where they have a key role in the development and maintenance of the functional circulatory system and provide paracrine support to surrounding non-vascular cells. Technical advances in the past 5 years in single-cell genomics and in in vivo genetic labelling have facilitated greater insights into endothelial cell development, plasticity and heterogeneity. These advances have also contributed to a new understanding of the timing of endothelial cell subtype differentiation and its relationship to the cell cycle. Identification of novel tissue-specific gene expression patterns in endothelial cells has led to the discovery of crucial signalling pathways and new interactions with other cell types that have key roles in both tissue maintenance and disease pathology. In this Review, we describe the latest findings in vascular endothelial cell development and diversity, which are often supported by large-scale, single-cell studies, and discuss the implications of these findings for vascular medicine. In addition, we highlight how techniques such as single-cell multimodal omics, which have become increasingly sophisticated over the past 2 years, are being utilized to study normal vascular physiology as well as functional perturbations in disease.
Collapse
Affiliation(s)
- Emily Trimm
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
45
|
Jevnikar K, Meglič A, Lapajne L, Logar M, Vidovič Valentinčič N, Globočnik Petrovič M, Jaki Mekjavić P. The Comparison of Retinal Microvascular Findings in Acute COVID-19 and 1-Year after Hospital Discharge Assessed with Multimodal Imaging-A Prospective Longitudinal Cohort Study. Int J Mol Sci 2023; 24:ijms24044032. [PMID: 36835445 PMCID: PMC9966689 DOI: 10.3390/ijms24044032] [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: 11/10/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023] Open
Abstract
This study aimed to quantify possible long-term impairment of the retinal microcirculation and microvasculature by reassessing a cohort of patients with acute COVID-19 without other known comorbidities one year after their discharge from the hospital. Thirty patients in the acute phase of COVID-19 without known systemic comorbidities were enrolled in this prospective longitudinal cohort study. Fundus photography, SS-OCT, and SS-OCTA using swept-source OCT (SS-OCT, Topcon DRI OCT Triton; Topcon Corp., Tokyo, Japan) were performed in the COVID-19 unit and 1-year after hospital discharge. The cohort's median age was 60 years (range 28-65) and 18 (60%) were male. Mean vein diameter (MVD) significantly decreased over time, from 134.8 μm in the acute phase to 112.4 μm at a 1-year follow-up (p < 0.001). A significantly reduced retinal nerve fiber layer (RNFL) thickness was observed at follow-up in the inferior quadrant of the inner ring (mean diff. 0.80 95% CI 0.01-1.60, p = 0.047) and inferior (mean diff. 1.56 95% CI 0.50-2.61, p < 0.001), nasal (mean diff. 2.21 95% CI 1.16-3.27, p < 0.001), and superior (mean diff. 1.69 95% CI 0.63-2.74, p < 0.001) quadrants of the outer ring. There were no statistically significant differences between the groups regarding vessel density of the superior and deep capillary plexuses. The transient dilatation of the retinal vessels in the acute phase of COVID-19, as well as RNFL thickness changes, could become a biomarker of angiopathy in patients with severe COVID-19.
Collapse
Affiliation(s)
- Kristina Jevnikar
- Department of Ophthalmology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Ophthalmology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Andrej Meglič
- Department of Ophthalmology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Luka Lapajne
- Department of Ophthalmology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Mateja Logar
- Department of Infectious Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Infectious Diseases, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Nataša Vidovič Valentinčič
- Department of Ophthalmology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Ophthalmology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mojca Globočnik Petrovič
- Department of Ophthalmology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Ophthalmology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Polona Jaki Mekjavić
- Department of Ophthalmology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Ophthalmology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
46
|
Han Z, Deng L, Chen S, Wang H, Huang Y. Zn 2+-Loaded adhesive bacterial cellulose hydrogel with angiogenic and antibacterial abilities for accelerating wound healing. BURNS & TRAUMA 2023; 11:tkac048. [PMID: 36751362 PMCID: PMC9897938 DOI: 10.1093/burnst/tkac048] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/09/2022] [Indexed: 02/05/2023]
Abstract
Background Wound healing is a process that requires angiogenesis and antibacterial activities and it remains a challenge for both experimental and clinical research worldwide. Zn2+ has been reported to be widely involved in angiogenesis and exerts antibacterial effects, making it suitable as a treatment to promote wound healing. Therefore Zn2+-loaded adhesive bacterial cellulose hydrogel was designed to observe its angiogenic and antibacterial abilities in the wound healing process. Methods The characterization, tensile strength, swelling behaviors and antibacterial activity of bacterial cellulose/polydopamine/zeolitic imidazolate framework-8 (BC/PDA/ZIF8) hydrogels were tested. Cell-Counting-Kit-8 (CCK8), transwell, tube formation and real time qunantitative PCR (qRT-PCR) assays were performed to evaluate the cell compatibility of BC/PDA/ZIF8 hydrogels in vitro. A full-thickness defect wound model and histological assays were used to evaluate the BC/PDA/ZIF8 hydrogels in vivo. Results The prepared BC/PDA/ZIF8 hydrogels exhibited suitable mechanical strength, excellent swelling properties, good tissue adhesion, efficient angiogenic and antibacterial effects and good performance as a physical barrier. In vivo experiments showed that the BC/PDA/ZIF8 hydrogels accelerated wound healing in a full-thickness defect wound model by stimulating angiogenesis. Conclusions This study proved that BC/PDA/ZIF8 hydrogels possess great potential for promoting satisfactory wound healing in full-thickness wound defects through antibacterial effects and improved cell proliferation, tissue formation, remodeling and re-epithelialization.
Collapse
Affiliation(s)
- Zhengzhe Han
- Department of Orthopedic Surgery, and Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, P.R. China
| | - Lili Deng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, P.R. China
| | - Shiyan Chen
- Correspondence Shiyan Chen, ; Huaping Wang, ; Yinjun Huang,
| | - Huaping Wang
- Correspondence Shiyan Chen, ; Huaping Wang, ; Yinjun Huang,
| | - Yinjun Huang
- Correspondence Shiyan Chen, ; Huaping Wang, ; Yinjun Huang,
| |
Collapse
|
47
|
Sun J, Lin Z, Liao Z, Wu Z, Li H, Wang H. Small extracellular vesicles derived from human adipose-derived stem cells regulate energetic metabolism through the activation of YAP/TAZ pathway facilitating angiogenesis. Cell Biol Int 2023; 47:451-466. [PMID: 36279478 DOI: 10.1002/cbin.11938] [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: 04/01/2022] [Revised: 08/30/2022] [Accepted: 10/11/2022] [Indexed: 01/17/2023]
Abstract
Recent studies have found small extracellular vesicles (sEVs) that are secreted from human adipose tissue-derived stem cells (hADSCs-sEVs) and contribute to angiogenesis. Glycolysis, the primary energetic pathway of vascular endothelial cells, plays a key role in the process of angiogenesis. However, hADSCs-sEVs' effects on energy metabolism within endothelial cells remain unclear. In our study, we found that hADSCs-sEVs restored glycolytic metabolism suppressed by 3-(pyridinyl)-1-(4-pyridinyl)-2-propen-1-one(3PO), a unique glycolytic inhibitor increasing the extracellular acidification rate (ECAR), oxygen consumption rate (OCR), glycolytic gene expression as well as pyruvate, lactate, and ATP production in HUVEC cells. In contrast, hADSCs-sEVs decreased PDH-E1α expression and acetyl-CoA production. The above results indicate that hADSCs-sEVs promote HUVEC angiogenesis via enhancing glycolysis and suppressing mitochondrial oxidative phosphorylation. Furthermore, we found that the YAP/TAZ pathway may play a key role in the effects hADSCs-sEVs have on HUVECs, thus, providing a promising approach for pro-angiogenesis-related regeneration.
Collapse
Affiliation(s)
- Jun Sun
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Zhengjie Lin
- Department of Plastic and Aesthetic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Zhi Liao
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Zhimin Wu
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Hao Li
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Hui Wang
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
48
|
Mirhaidari GJ, Barker JC, Breuer CK, Reinhardt JW. Implanted Tissue-Engineered Vascular Graft Cell Isolation with Single-Cell RNA Sequencing Analysis. Tissue Eng Part C Methods 2023; 29:72-84. [PMID: 36719780 PMCID: PMC9968626 DOI: 10.1089/ten.tec.2022.0189] [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: 11/12/2022] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
The advent of single-cell RNA sequencing (scRNA-Seq) has brought with it the ability to gain greater insights into the cellular composition of tissues and heterogeneity in gene expression within specific cell types. For tissue-engineered blood vessels, this is particularly impactful to better understand how neotissue forms and remodels into tissue resembling a native vessel. A notable challenge, however, is the ability to separate cells from synthetic biomaterials to generate high-quality single-cell suspensions to interrogate the cellular composition of our tissue-engineered vascular grafts (TEVGs) during active remodeling in situ. We present here a simple, commercially available approach to separate cells within our TEVG from the residual scaffold for downstream use in a scRNA-Seq workflow. Utilizing this method, we identified the cell populations comprising explanted TEVGs and compared these with results from immunohistochemical analysis. The process began with explanted TEVGs undergoing traditional mechanical and enzymatic dissociation to separate cells from scaffold and extracellular matrix proteins. Magnetically labeled antibodies targeting murine origin cells were incubated with enzymatic digests of TEVGs containing cells and scaffold debris in suspension allowing for separation by utilizing a magnetic separator column. Single-cell suspensions were processed through 10 × Genomics and data were analyzed utilizing R to generate cell clusters. Expression data provided new insights into a diverse composition of phenotypically unique subclusters within the fibroblast, macrophage, smooth muscle cell, and endothelial cell populations contributing to the early neotissue remodeling stages of TEVGs. These populations were correlated qualitatively and quantitatively with immunohistochemistry highlighting for the first time the potential of scRNA-Seq to provide exquisite detail into the host cellular response to an implanted TEVG. These results additionally demonstrate magnetic cell isolation is an effective method for generating high-quality cell suspensions for scRNA-Seq. While this method was utilized for our group's TEVGs, it has broader applications to other implantable materials that use biodegradable synthetic materials as part of scaffold composition. Impact statement Single-cell RNA sequencing is an evolving technology with the ability to provide detailed information on the cellular composition of remodeling biomaterials in vivo. This present work details an effective approach for separating nondegraded biomaterials from cells for downstream RNA-sequencing analysis. We applied this method to implanted tissue-engineered vascular grafts and for the first time describe the cellular composition of the remodeling graft at a single-cell gene expression level. While this method was effective in our scaffold, it has broad applicability to other implanted biomaterials that necessitate separation of cell from residual scaffold materials for single-cell RNA sequencing.
Collapse
Affiliation(s)
- Gabriel J.M. Mirhaidari
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Jenny C. Barker
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Plastic and Reconstructive Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Christopher K. Breuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - James W. Reinhardt
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| |
Collapse
|
49
|
Bloom SI, Islam MT, Lesniewski LA, Donato AJ. Mechanisms and consequences of endothelial cell senescence. Nat Rev Cardiol 2023; 20:38-51. [PMID: 35853997 PMCID: PMC10026597 DOI: 10.1038/s41569-022-00739-0] [Citation(s) in RCA: 189] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 12/15/2022]
Abstract
Endothelial cells are located at the crucial interface between circulating blood and semi-solid tissues and have many important roles in maintaining systemic physiological function. The vascular endothelium is particularly susceptible to pathogenic stimuli that activate tumour suppressor pathways leading to cellular senescence. We now understand that senescent endothelial cells are highly active, secretory and pro-inflammatory, and have an aberrant morphological phenotype. Moreover, endothelial senescence has been identified as an important contributor to various cardiovascular and metabolic diseases. In this Review, we discuss the consequences of endothelial cell exposure to damaging stimuli (haemodynamic forces and circulating and endothelial-derived factors) and the cellular and molecular mechanisms that induce endothelial cell senescence. We also discuss how endothelial cell senescence causes arterial dysfunction and contributes to clinical cardiovascular diseases and metabolic disorders. Finally, we summarize the latest evidence on the effect of eliminating senescent endothelial cells (senolysis) and identify important remaining questions to be addressed in future studies.
Collapse
Affiliation(s)
- Samuel I Bloom
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Md Torikul Islam
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Lisa A Lesniewski
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
- Veterans Affairs Medical Center-Salt Lake City, Geriatric Research Education and Clinical Center, Salt Lake City, UT, USA
| | - Anthony J Donato
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA.
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA.
- Veterans Affairs Medical Center-Salt Lake City, Geriatric Research Education and Clinical Center, Salt Lake City, UT, USA.
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
50
|
Wakabayashi T, Naito H. Cellular heterogeneity and stem cells of vascular endothelial cells in blood vessel formation and homeostasis: Insights from single-cell RNA sequencing. Front Cell Dev Biol 2023; 11:1146399. [PMID: 37025170 PMCID: PMC10070846 DOI: 10.3389/fcell.2023.1146399] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
Vascular endothelial cells (ECs) that constitute the inner surface of blood vessels are essential for new vessel formation and organ homeostasis. ECs display remarkable phenotypic heterogeneity across different organs and the vascular tree during angiogenesis and homeostasis. Recent advances in single cell RNA sequencing (scRNA-seq) technologies have allowed a new understanding of EC heterogeneity in both mice and humans. In particular, scRNA-seq has identified new molecular signatures for arterial, venous and capillary ECs in different organs, as well as previously unrecognized specialized EC subtypes, such as the aerocytes localized in the alveolar capillaries of the lung. scRNA-seq has also revealed the gene expression profiles of specialized tissue-resident EC subtypes that are capable of clonal expansion and contribute to adult angiogenesis, a process of new vessel formation from the pre-existing vasculature. These specialized tissue-resident ECs have been identified in various different mouse tissues, including aortic endothelium, liver, heart, lung, skin, skeletal muscle, retina, choroid, and brain. Transcription factors and signaling pathways have also been identified in the specialized tissue-resident ECs that control angiogenesis. Furthermore, scRNA-seq has also documented responses of ECs in diseases such as cancer, age-related macular degeneration, Alzheimer's disease, atherosclerosis, and myocardial infarction. These new findings revealed by scRNA-seq have the potential to provide new therapeutic targets for different diseases associated with blood vessels. In this article, we summarize recent advances in the understanding of the vascular endothelial cell heterogeneity and endothelial stem cells associated with angiogenesis and homeostasis in mice and humans, and we discuss future prospects for the application of scRNA-seq technology.
Collapse
Affiliation(s)
- Taku Wakabayashi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
- Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, United States
- *Correspondence: Taku Wakabayashi, ; Hisamichi Naito,
| | - Hisamichi Naito
- Department of Vascular Physiology, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa, Japan
- *Correspondence: Taku Wakabayashi, ; Hisamichi Naito,
| |
Collapse
|