1
|
Al-Hakeim HK, Twaij BAAR, Al-Naqeeb TH, Moustafa SR, Maes M. Neuronal damage and inflammatory biomarkers are associated with the affective and chronic fatigue-like symptoms due to end-stage renal disease. J Affect Disord 2024; 347:220-229. [PMID: 38007104 DOI: 10.1016/j.jad.2023.11.078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Many biochemical, immunological, and neuropsychiatric changes are associated with end-stage renal disease (ESRD). Neuronal damage biomarkers such as glial fibrillary acidic protein (GFAP), neurofilament light chain (NFL), S100 calcium-binding protein B (S100B), ionized calcium-binding adaptor molecule-1 (IBA1), and myelin basic protein (MBP) are among the less-studied biomarkers of ESRD. AIM We examined the associations between these neuro-axis biomarkers, inflammatory biomarkers, e.g., C-reactive protein (CRP), interleukin (IL-6), IL-10, and zinc, copper, and neuropsychiatric symptoms due to ERSD. METHODS ELISA techniques were used to measure serum levels of neuronal damage biomarkers in 70 ESRD patients, and 46 healthy controls. RESULTS ESRD patients have higher scores of depression, anxiety, fatigue, and physiosomatic symptoms than healthy controls. Aberrations in kidney function tests and the number of dialysis interventions are associated with the severity of depression, anxiety, fibro-fatigue and physiosomatic symptoms, peripheral inflammation, nestin, and NFL. Serum levels of neuronal damage biomarkers (NFL, MBP, and nestin), CRP, and interleukin (IL)-10 are elevated, and serum zinc is decreased in ESRD patients as compared with controls. The neuronal damage biomarkers NFL, nestin, S100B and MBP are associated with the severity of one or more neuropsychiatric symptom domains. Around 50 % of the variance in the neuropsychiatric symptoms is explained by NFL, nestin, S00B, copper, and an inflammatory index. CONCLUSIONS The severity of renal dysfunction and/or the number of dialysis interventions may induce peripheral inflammation and, consequently, neurotoxicity to intermediate filament proteins, astrocytes, and the blood-brain barrier, leading to the neuropsychiatric symptoms of ESRD.
Collapse
Affiliation(s)
| | | | - Tabarek Hadi Al-Naqeeb
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Shatha Rouf Moustafa
- Clinical Analysis Department, College of Pharmacy, Hawler Medical University, Havalan City, Erbil, Iraq
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University of Plovdiv, Plovdiv, Bulgaria; Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China.
| |
Collapse
|
2
|
Al-hakeim HK, Al-raheem Twaij BA, Al-naqeeb TH, Moustafa SR, Maes M. Neuronal damage and inflammatory biomarkers are associated with the affective and chronic fatigue-like symptoms due to end-stage renal disease.. [DOI: 10.1101/2023.05.03.23289492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
AbstractBackgroundMany biochemical, immunological, and neuropsychiatric changes are associated with end-stage renal disease (ESRD). Neuronal damage biomarkers such as glial fibrillary acidic protein (GFAP), neurofilament light chain (NFL), S100 calcium-binding protein B (S100B), ionized calcium-binding adaptor molecule-1 (IBA1), and myelin basic protein (MBP) are among the less-studied biomarkers of ESRD.AimWe examined the associations between these neuro-axis biomarkers, inflammatory biomarkers, e.g., C-reactive protein (CRP), interleukin (IL-6), IL-10, and zinc, copper, and neuropsychiatric symptoms due to ERSD.MethodsELISA techniques were used to measure serum levels of neuronal damage biomarkers in 70 ESRD patients, and 46 healthy controls.ResultsESRD patients have higher scores of depression, anxiety, fatigue, and physiosomatic symptoms than healthy controls. Aberrations in kidney function tests and the number of dialysis interventions are associated with the severity of depression, anxiety, fibro-fatigue and physiosomatic symptoms, peripheral inflammation, nestin, and NFL. Serum levels of neuronal damage biomarkers (NFL, MBP, and nestin), CRP, and interleukin (IL)-10 are elevated, and serum zinc is decreased in ESRD patients as compared with controls. The neuronal damage biomarkers NFL, nestin, S100B and MBP are associated with the severity of one or more neuropsychiatric symptom domains. Around 50% of the variance in the neuropsychiatric symptoms is explained by NFL, nestin, S00B, copper, and an inflammatory index.ConclusionsThe severity of renal dysfunction and/or the number of dialysis interventions may induce peripheral inflammation and, consequently, neurotoxicity to intermediate filament proteins, astrocytes, and the blood-brain barrier, leading to the neuropsychiatric symptoms of ESRD.
Collapse
|
3
|
Derhambakhsh S, Mohammadi J, Shokrgozar MA, Rabbani H, Sadeghi N, Nekounam H, Mohammadi S, Lee KB, Khakbiz M. Investigation of electrical stimulation on phenotypic vascular smooth muscle cells differentiation in tissue-engineered small-diameter vascular graft. Tissue Cell 2023; 81:101996. [PMID: 36657256 DOI: 10.1016/j.tice.2022.101996] [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: 07/18/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
In the development of vascular tissue engineering, particularly in the case of small diameter vessels, one of the key obstacles is the blockage of these veins once they enter the in vivo environment. One of the contributing factors to this problem is the aberrant proliferation and migration of vascular smooth muscle cells (VSMCs) from the media layer of the artery to the interior of the channel. Two distinct phenotypes have been identified for smooth muscle cells, namely synthetic and contractile. Since the synthetic phenotype plays an essential role in the unusual growth and migration, the aim of this study was to convert the synthetic phenotype into the contractile one, which is a solution to prevent the abnormal growth of VSMCs. To achieve this goal, these cells were subjected to electrical signals, using a 1000 μA sinusoidal stimulation at 10 Hz for four days, with 20 min duration per 24 h. The morphological transformations and changes in the expression of vimentin, nestin, and β-actin proteins were then studied using ICC and flow cytometry assays. Also, the expression of VSMC specific markers such as smooth muscle myosin heavy chain (SMMHC) and smooth muscle alpha-actin (α-SMA) were evaluated using RT-PCR test. In the final phase of this study, the sheep decellularized vessel was employed as a scaffold for seeding these cells. Based on the results, electrical stimulation resulted in some morphological alterations in VSMCs. Furthermore, the observed reductions in the expression levels of vimentin, nestin and β-actin proteins and increase in the expression of SMMHC and α-SMA markers showed that it is possible to convert the synthetic phenotype to the contractile one using the studied regime of electrical stimulation. Finally, it can be concluded that electrical stimulation can significantly affect the phenotype of VSMCs, as demonstrated in this study.
Collapse
Affiliation(s)
- Sara Derhambakhsh
- Division of Biomedical Engineering, Department of Life Science, Faculty of New Sciences and Technologies, University of Tehran, Tehran 439957131, Iran
| | - Javad Mohammadi
- Division of Biomedical Engineering, Department of Life Science, Faculty of New Sciences and Technologies, University of Tehran, Tehran 439957131, Iran.
| | | | - Hodjattallah Rabbani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Niloufar Sadeghi
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Houra Nekounam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Sotoudeh Mohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Mehrdad Khakbiz
- Division of Biomedical Engineering, Department of Life Science, Faculty of New Sciences and Technologies, University of Tehran, Tehran 439957131, Iran.
| |
Collapse
|
4
|
Shi C, Zhang K, Zhao Z, Wang Y, Xu H, Wei W. Correlation between stem cell molecular phenotype and atherosclerotic plaque neointima formation and analysis of stem cell signal pathways. Front Cell Dev Biol 2023; 11:1080563. [PMID: 36711040 PMCID: PMC9877345 DOI: 10.3389/fcell.2023.1080563] [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: 10/26/2022] [Accepted: 01/02/2023] [Indexed: 01/14/2023] Open
Abstract
Vascular stem cells exist in the three-layer structure of blood vessel walls and play an indispensable role in angiogenesis under physiological conditions and vascular remodeling under pathological conditions. Vascular stem cells are mostly quiescent, but can be activated in response to injury and participate in endothelial repair and neointima formation. Extensive studies have demonstrated the differentiation potential of stem/progenitor cells to repair endothelium and participate in neointima formation during vascular remodeling. The stem cell population has markers on the surface of the cells that can be used to identify this cell population. The main positive markers include Stem cell antigen-1 (Sca1), Sry-box transcription factor 10 (SOX10). Stromal cell antigen 1 (Stro-1) and Stem cell growth factor receptor kit (c-kit) are still controversial. Different parts of the vessel have different stem cell populations and multiple markers. In this review, we trace the role of vascular stem/progenitor cells in the progression of atherosclerosis and neointima formation, focusing on the expression of stem cell molecular markers that occur during neointima formation and vascular repair, as well as the molecular phenotypic changes that occur during differentiation of different stem cell types. To explore the correlation between stem cell molecular markers and atherosclerotic diseases and neointima formation, summarize the differential changes of molecular phenotype during the differentiation of stem cells into smooth muscle cells and endothelial cells, and further analyze the signaling pathways and molecular mechanisms of stem cells expressing different positive markers participating in intima formation and vascular repair. Summarizing the limitations of stem cells in the prevention and treatment of atherosclerotic diseases and the pressing issues that need to be addressed, we provide a feasible scheme for studying the signaling pathways of vascular stem cells involved in vascular diseases.
Collapse
Affiliation(s)
- Chuanxin Shi
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kefan Zhang
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenyu Zhao
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifan Wang
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haozhe Xu
- Department of Biotherapy, Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wei
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Wei Wei,
| |
Collapse
|
5
|
Kirkton RD, Santiago-Maysonet M, Lawson JH, Tente WE, Dahl SLM, Niklason LE, Prichard HL. Bioengineered human acellular vessels recellularize and evolve into living blood vessels after human implantation. Sci Transl Med 2020; 11:11/485/eaau6934. [PMID: 30918113 DOI: 10.1126/scitranslmed.aau6934] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 03/06/2019] [Indexed: 12/13/2022]
Abstract
Traditional vascular grafts constructed from synthetic polymers or cadaveric human or animal tissues support the clinical need for readily available blood vessels, but often come with associated risks. Histopathological evaluation of these materials has shown adverse host cellular reactions and/or mechanical degradation due to insufficient or inappropriate matrix remodeling. We developed an investigational bioengineered human acellular vessel (HAV), which is currently being studied as a hemodialysis conduit in patients with end-stage renal disease. In rare cases, small samples of HAV were recovered during routine surgical interventions and used to examine the temporal and spatial pattern of the host cell response to the HAV after implantation, from 16 to 200 weeks. We observed a substantial influx of alpha smooth muscle actin (αSMA)-expressing cells into the HAV that progressively matured and circumferentially aligned in the HAV wall. These cells were supported by microvasculature initially formed by CD34+/CD31+ cells in the neoadventitia and later maintained by CD34-/CD31+ endothelial cells in the media and lumen of the HAV. Nestin+ progenitor cells differentiated into either αSMA+ or CD31+ cells and may contribute to early recellularization and self-repair of the HAV. A mesenchymal stem cell-like CD90+ progenitor cell population increased in number with duration of implantation. Our results suggest that host myogenic, endothelial, and progenitor cell repopulation of HAVs transforms these previously acellular vessels into functional multilayered living tissues that maintain blood transport and exhibit self-healing after cannulation injury, effectively rendering these vessels like the patient's own blood vessel.
Collapse
Affiliation(s)
| | | | - Jeffrey H Lawson
- Humacyte Inc., Durham, NC 27713, USA.,Departments of Surgery and Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | - Laura E Niklason
- Humacyte Inc., Durham, NC 27713, USA.,Departments of Anesthesiology and Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | | |
Collapse
|
6
|
Møller S, Jacobsen JCB, Braunstein TH, Holstein-Rathlou NH, Sorensen CM. Influence of connexin45 on renal autoregulation. Am J Physiol Renal Physiol 2020; 318:F732-F740. [DOI: 10.1152/ajprenal.00185.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Renal autoregulation is mediated by the myogenic response and tubuloglomerular feedback (TGF) working in concert to maintain renal blood flow and glomerular filtration rate despite fluctuations in renal perfusion pressure. Intercellular communication through gap junctions may play a role in renal autoregulation. We examine if one of the building blocks in gap junctions, connexin45 (Cx45), which is expressed in vascular smooth muscle cells, has an influence on renal autoregulatory efficiency. The isolated perfused juxtamedullary nephron preparation was used to measure afferent arteriolar diameter changes in response to acute changes in renal perfusion pressure. In segmental arteries, pressure myography was used to study diameter changes in response to pressure changes. Wire myography was used to study vasoconstrictor and vasodilator responses. A mathematical model of the vascular wall was applied to interpret experimental data. We found a significant reduction in the afferent arteriolar constriction in response to acute pressure increases in Cx45 knockout (KO) mice compared with wild-type (WT) mice. Abolition of TGF caused a parallel upward shift in the autoregulation curve of WT animals but had no effect in KO animals, which is compatible with TGF providing a basal tonic contribution in afferent arterioles whereas Cx45 KO animals were functionally papillectomized. Analysis showed a shift toward lower stress sensitivity in afferent arterioles from Cx45 KO animals, indicating that the absence of Cx45 may also affect myogenic properties. Finally, loss of Cx45 in vascular smooth muscle cells appeared to associate with a change in both structure and passive properties of the vascular wall.
Collapse
Affiliation(s)
- Sophie Møller
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Christian Brings Jacobsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas H. Braunstein
- Core Facility for Integrated Microscopy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels-Henrik Holstein-Rathlou
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte M. Sorensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Witt E, Lorenz M, Völker U, Stangl K, Hammer E, Stangl V. Sex-specific differences in the intracellular proteome of human endothelial cells from dizygotic twins. J Proteomics 2019; 201:48-56. [PMID: 30951907 DOI: 10.1016/j.jprot.2019.03.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/11/2019] [Accepted: 03/27/2019] [Indexed: 12/19/2022]
Abstract
Differences between men and women are being continuously identified in many human diseases. The underlying reasons are not yet fully understood. Beside the influence of endogenous hormones and life style, intrinsic sex-specific dimorphisms at the cellular level may also play a role. HUVECs from twin pairs of opposite sex provide an excellent tool to address the question of sex-specific differences at the molecular level. We compared for the first time protein levels of male and female HUVECs from dizygotic twins using a proteomic approach. To investigate differences under basal and stress conditions, cells were either left untreated or wounded and serum starved for different time points. Approximately 10% of all proteins monitored showed significant sexual dimorphisms in their level under the different conditions tested. The majority of the proteins displayed a higher abundance in female cells. The magnitude of the difference in protein levels between male and female cells was rather small. The most prominent differences throughout all conditions were observed for several X-chromosome encoded proteins with higher levels in female (UBA1, HDHD1) or in male cells (G6PD). Proteins involved in basic cellular processes, such as gene expression and translation (e.g. HMGN1, SRP54) displayed sex-specific levels in particular conditions only. SIGNIFICANCE: This study provides novel insights into sexual dimorphic protein levels in HUVECs from twin pairs of the opposite sex. The findings identify proteins with sex-specific differences in their levels under different cell culture conditions. The study also highlights the presence of X-chromosome encoded proteins escaping X-chromosomal inactivation. The results emphasize the need to consider the cellular sex of male and female HUVECs in in vitro experiments.
Collapse
Affiliation(s)
- Eric Witt
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany
| | - Mario Lorenz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany
| | - Karl Stangl
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany; DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany.
| | - Verena Stangl
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| |
Collapse
|
8
|
Sabharwal R, Mason BN, Kuburas A, Abboud FM, Russo AF, Chapleau MW. Increased receptor activity-modifying protein 1 in the nervous system is sufficient to protect against autonomic dysregulation and hypertension. J Cereb Blood Flow Metab 2019; 39:690-703. [PMID: 29297736 PMCID: PMC6446426 DOI: 10.1177/0271678x17751352] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Calcitonin gene-related peptide (CGRP) can cause migraines, yet it is also a potent vasodilator that protects against hypertension. Given the emerging role of CGRP-targeted antibodies for migraine prevention, an important question is whether the protective actions of CGRP are mediated by vascular or neural CGRP receptors. To address this, we have characterized the cardiovascular phenotype of transgenic nestin/hRAMP1 mice that have selective elevation of a CGRP receptor subunit in the nervous system, human receptor activity-modifying protein 1 (hRAMP1). Nestin/hRAMP1 mice had relatively little hRAMP1 RNA in blood vessels and intravenous injection of CGRP caused a similar blood pressure decrease in transgenic and control mice. At baseline, nestin/hRAMP1 mice exhibited similar mean arterial pressure, heart rate, baroreflex sensitivity, and sympathetic vasomotor tone as control mice. We previously reported that expression of hRAMP1 in all tissues favorably improved autonomic regulation and attenuated hypertension induced by angiotensin II (Ang II). Similarly, in nestin/hRAMP1 mice, hypertension caused by Ang II or phenylephrine was greatly attenuated, and associated autonomic dysregulation and increased sympathetic vasomotor tone were diminished or abolished. We conclude that increased expression of neuronal CGRP receptors is sufficient to induce a protective change in cardiovascular autonomic regulation with implications for migraine therapy.
Collapse
Affiliation(s)
- Rasna Sabharwal
- 1 Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Bianca N Mason
- 2 Molecular and Cell Biology Program, University of Iowa, Iowa City, IA, USA
| | - Adisa Kuburas
- 3 Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Francois M Abboud
- 1 Department of Internal Medicine, University of Iowa, Iowa City, IA, USA.,3 Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Andrew F Russo
- 2 Molecular and Cell Biology Program, University of Iowa, Iowa City, IA, USA.,3 Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.,4 Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,5 Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Mark W Chapleau
- 1 Department of Internal Medicine, University of Iowa, Iowa City, IA, USA.,3 Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.,5 Veterans Affairs Medical Center, Iowa City, IA, USA
| |
Collapse
|
9
|
Leong ZP, Hikasa Y. Effects of toceranib compared with sorafenib on monocrotaline-induced pulmonary arterial hypertension and cardiopulmonary remodeling in rats. Vascul Pharmacol 2018; 110:31-41. [PMID: 30071297 DOI: 10.1016/j.vph.2018.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/04/2018] [Accepted: 07/29/2018] [Indexed: 02/07/2023]
Abstract
Sorafenib reverses pulmonary arterial hypertension (PAH) and cardiopulmonary remodeling (CPR), but the effects of toceranib are unknown. This study investigated anti-remodeling effects and determined optimal doses of toceranib and sorafenib on monocrotaline (MCT)-induced PAH and CPR in rats. MCT-treated rats were orally treated with a 14-day course of sorafenib (10, 30, or 100 mg/kg), toceranib (1, 3, or 10 mg/kg), or water. Both sorafenib and toceranib significantly reversed the right ventricular (RV) hypertrophy at 10 mg/kg, but only sorafenib significantly improved the RV systolic and mean pressures. Sorafenib significantly normalized the B-type natriuretic peptide mRNA level of the RV and increased the non-muscularized pulmonary artery percentage. However, these effects were only observed at the highest toceranib dose, and neither toceranib dose reduced the fully muscularized pulmonary artery percentage. Further, the inhibition on vascular endothelial growth factor (VEGF) signaling was stronger in sorafenib than in toceranib. Besides the stronger inhibition on mitogen-activated protein kinase signaling, the greater reversal ability of sorafenib may be also due to the simultaneous blockade on the C-X-C chemokine receptor type 4 and autophagy induction. Toceranib insignificantly reversed CPR, and a high-dose therapy did not improve the RV hemodynamic outcomes. Sorafenib significantly reversed CPR, and a low-dose sorafenib therapy may be a suitable therapeutic agent for PAH.
Collapse
Affiliation(s)
- Zi Ping Leong
- The United Graduate School of Veterinary Science, Yamaguchi University, 1677-1, Yoshida, Yamaguchi 753-8515, Japan
| | - Yoshiaki Hikasa
- The United Graduate School of Veterinary Science, Yamaguchi University, 1677-1, Yoshida, Yamaguchi 753-8515, Japan; Joint Department of Veterinary Medicine, Laboratory of Veterinary Internal Medicine, Tottori University, Tottori 680-8550, Japan.
| |
Collapse
|
10
|
He S, Lin J, Lin L, Xu Y, Feng J. Shikonin‑mediated inhibition of nestin affects hypoxia‑induced proliferation of pulmonary artery smooth muscle cells. Mol Med Rep 2018; 18:3476-3482. [PMID: 30066896 DOI: 10.3892/mmr.2018.9333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/21/2018] [Indexed: 11/06/2022] Open
Abstract
The imbalance between the proliferation and apoptosis of pulmonary artery smooth muscle cells (PASMCs) is of importance in pulmonary vascular remodeling. Shikonin, a naphthoquinone compound extracted from the Chinese medicinal herb Lithospermum erythrorhizon, inhibits the proliferation of rat smooth muscle cells (SMCs). The present study was designed to investigate the effects of shikonin on the proliferation of rat PASMCs and the possible mechanisms involved. Rat PASMCs were cultured under the following five treatment conditions: Normal control; hypoxia for 24 h; hypoxia + 1 µM shikonin for 24 h; hypoxia + 2 µM shikonin for 24 h; and hypoxia + 4 µM shikonin for 24 h. The viability of PASMCs was measured using the Cell Counting Kit‑8 assay, the mRNA expression of nestin (NES) in each group was measured by reverse transcription‑polymerase chain reaction and the protein expression of NES was measured by western blotting. The proliferation of hypoxic PASMCs transfected with NES‑specific small interfering (si)RNA decreased compared with the non‑transfected group. These results indicated that hypoxia induced the proliferation of PASMCs through the enhancement of NES expression. The treatment of hypoxic PASMCs with shikonin resulted in a significant downregulation of NES expression and the inhibition of PASMC proliferation.
Collapse
Affiliation(s)
- Susu He
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, P.R. China
| | - Jian Lin
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, P.R. China
| | - Ling Lin
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, P.R. China
| | - Youzu Xu
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, P.R. China
| | - Jiaxi Feng
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, P.R. China
| |
Collapse
|
11
|
Reckmann AN, Tomczyk CUM, Davidoff MS, Michurina TV, Arnhold S, Müller D, Mietens A, Middendorff R. Nestin in the epididymis is expressed in vascular wall cells and is regulated during postnatal development and in case of testosterone deficiency. PLoS One 2018; 13:e0194585. [PMID: 29874225 PMCID: PMC5991371 DOI: 10.1371/journal.pone.0194585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 03/06/2018] [Indexed: 12/24/2022] Open
Abstract
Vascular smooth muscle cells (SMCs), distinguished by the expression of the neuronal stem cell marker nestin, may represent stem cell-like progenitor cells in various organs including the testis. We investigated epididymal tissues of adult nestin-GFP mice, rats after Leydig cell depletion via ethane dimethane sulfonate (EDS), rats and mice during postnatal development and human tissues. By use of Clarity, a histochemical method to illustrate a three-dimensional picture, we could demonstrate nestin-GFP positive cells within the vascular network. We localized nestin in the epididymis in proliferating vascular SMCs by colocalization with both smooth muscle actin and PCNA, and it was distinct from CD31-positive endothelial cells. The same nestin localization was found in the human epididymis. However, nestin was not found in SMCs of the epididymal duct. Nestin expression is high during postnatal development of mouse and rat and down-regulated towards adulthood when testosterone levels increase. Nestin increases dramatically in rats after Leydig cell ablation with EDS and subsequently low testosterone levels. Interestingly, during this period, the expression of androgen receptor in the epididymis is low and increases until nestin reaches normal levels of adulthood. Here we show that nestin, a common marker for neuronal stem cells, is also expressed in the vasculature of the epididymis. Our results give new insights into the yet underestimated role of proliferating nestin-expressing vascular SMCs during postnatal development and repair of the epididymis.
Collapse
Affiliation(s)
- Ansgar N Reckmann
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Claudia U M Tomczyk
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Michail S Davidoff
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tatyana V Michurina
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States of America
- Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States of America
- Moscow Institute of Physics and Technology, Moscow, Russia
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Dieter Müller
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Andrea Mietens
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Ralf Middendorff
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
12
|
Calderone A. The Biological Role of Nestin (+)-Cells in Physiological and Pathological Cardiovascular Remodeling. Front Cell Dev Biol 2018; 6:15. [PMID: 29492403 PMCID: PMC5817075 DOI: 10.3389/fcell.2018.00015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 01/31/2018] [Indexed: 01/02/2023] Open
Abstract
The intermediate filament protein nestin was identified in diverse populations of cells implicated in cardiovascular remodeling. Cardiac resident neural progenitor/stem cells constitutively express nestin and following an ischemic insult migrate to the infarct region and participate in angiogenesis and neurogenesis. A modest number of normal adult ventricular fibroblasts express nestin and the intermediate filament protein is upregulated during the progression of reparative and reactive fibrosis. Nestin depletion attenuates cell cycle re-entry suggesting that increased expression of the intermediate filament protein in ventricular fibroblasts may represent an activated phenotype accelerating the biological impact during fibrosis. Nestin immunoreactivity is absent in normal adult rodent ventricular cardiomyocytes. Following ischemic damage, the intermediate filament protein is induced in a modest population of pre-existing adult ventricular cardiomyocytes bordering the peri-infarct/infarct region and nestin(+)-ventricular cardiomyocytes were identified in the infarcted human heart. The appearance of nestin(+)-ventricular cardiomyocytes post-myocardial infarction (MI) recapitulates an embryonic phenotype and depletion of the intermediate filament protein inhibits cell cycle re-entry. Recruitment of the serine/threonine kinase p38 MAPK secondary to an overt inflammatory response after an ischemic insult may represent a seminal event limiting the appearance of nestin(+)-ventricular cardiomyocytes and concomitantly suppressing cell cycle re-entry. Endothelial and vascular smooth muscle cells (VSMCs) express nestin and upregulation of the intermediate filament protein may directly contribute to vascular remodeling. This review will highlight the biological role of nestin(+)-cells during physiological and pathological remodeling of the heart and vasculature and discuss the phenotypic advantage attributed to the intermediate filament protein.
Collapse
Affiliation(s)
- Angelino Calderone
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada.,Montreal Heart Institute, Montréal, QC, Canada
| |
Collapse
|
13
|
Samelson EJ, Demissie S, Cupples LA, Zhang X, Xu H, Liu CT, Boyd SK, McLean RR, Broe KE, Kiel DP, Bouxsein ML. Diabetes and Deficits in Cortical Bone Density, Microarchitecture, and Bone Size: Framingham HR-pQCT Study. J Bone Miner Res 2018; 33:54-62. [PMID: 28929525 PMCID: PMC5771832 DOI: 10.1002/jbmr.3240] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 06/21/2017] [Accepted: 07/05/2017] [Indexed: 12/14/2022]
Abstract
Older adults with type 2 diabetes (T2D) tend to have normal or greater areal bone mineral density (aBMD), as measured by DXA, than those who do not have diabetes (non-T2D). Yet risk of fracture is higher in T2D, including 40% to 50% increased hip fracture risk. We used HR-pQCT to investigate structural mechanisms underlying skeletal fragility in T2D. We compared cortical and trabecular bone microarchitecture, density, bone area, and strength in T2D and non-T2D. In secondary analyses we evaluated whether associations between T2D and bone measures differed according to prior fracture, sex, and obesity. Participants included 1069 members of the Framingham Study, who attended examinations in 2005 to 2008 and underwent HR-pQCT scanning in 2012 to 2015. Mean age was 64 ± 8 years (range, 40 to 87 years), and 12% (n = 129) had T2D. After adjustment for age, sex, weight, and height, T2D had lower cortical volumetric BMD (vBMD) (p < 0.01), higher cortical porosity (p = 0.02), and smaller cross-sectional area (p = 0.04) at the tibia, but not radius. Trabecular indices were similar or more favorable in T2D than non-T2D. Associations between T2D and bone measures did not differ according to sex or obesity status (all interaction p > 0.05); however, associations did differ in those with a prior fracture and those with no history of fracture. Specifically, cortical vBMD at the tibia and cortical thickness at the radius were lower in T2D than non-T2D, but only among those individuals with a prior fracture. Cortical porosity at the radius was higher in T2D than non-T2D, but only among those who did not have a prior fracture. Findings from this large, community-based study of older adults suggest that modest deterioration in cortical bone and reductions in bone area may characterize diabetic bone disease in older adults. Evaluation of these deficits as predictors of fracture in T2D is needed to develop prevention strategies in this rapidly increasing population of older adults. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Elizabeth J. Samelson
- Institute for Aging Research, Hebrew Senior Life, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Gerontology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Serkalem Demissie
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Xiaochun Zhang
- Institute for Aging Research, Hebrew Senior Life, Boston, MA, USA
| | - Hanfei Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Steven K. Boyd
- McCaig Institute for Bone and Joint Health, University of Calgary, Canada
| | - Robert R. McLean
- Institute for Aging Research, Hebrew Senior Life, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Gerontology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kerry E. Broe
- Institute for Aging Research, Hebrew Senior Life, Boston, MA, USA
| | - Douglas P. Kiel
- Institute for Aging Research, Hebrew Senior Life, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Gerontology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Mary L. Bouxsein
- Beth Israel Deaconess Medical Center, Center for Advanced Orthopedic Studies, Boston, MA, USA
| |
Collapse
|
14
|
Reversal effects of low-dose imatinib compared with sunitinib on monocrotaline-induced pulmonary and right ventricular remodeling in rats. Vascul Pharmacol 2018; 100:41-50. [DOI: 10.1016/j.vph.2017.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/21/2017] [Accepted: 10/29/2017] [Indexed: 12/14/2022]
|
15
|
Hertig V, Matos-Nieves A, Garg V, Villeneuve L, Mamarbachi M, Caland L, Calderone A. Nestin expression is dynamically regulated in cardiomyocytes during embryogenesis. J Cell Physiol 2017; 233:3218-3229. [PMID: 28834610 DOI: 10.1002/jcp.26165] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/17/2017] [Accepted: 08/22/2017] [Indexed: 12/30/2022]
Abstract
The transcriptional factors implicated in the expression of the intermediate filament protein nestin in cardiomyocytes during embryogenesis remain undefined. In the heart of 9,5-10,5 day embryonic mice, nestin staining was detected in atrial and ventricular cardiomyocytes and a subpopulation co-expressed Tbx5. At later stages of development, nestin immunoreactivity in cardiomyocytes gradually diminished and was absent in the heart of 17,5 day embryonic mice. In the heart of wild type 11,5 day embryonic mice, 54 ± 7% of the trabeculae expressed nestin and the percentage was significantly increased in the hearts of Tbx5+/- and Gata4+/- embryos. The cell cycle protein Ki67 and transcriptional coactivator Yap-1 were still prevalent in the nucleus of nestin(+) -cardiomyocytes identified in the heart of Tbx5+/- and Gata4+/- embryonic mice. Phorbol 12,13-dibutyrate treatment of neonatal rat ventricular cardiomyocytes increased Yap-1 phosphorylation and co-administration of the p38 MAPK inhibitor SB203580 led to significant dephosphorylation. Antagonism of dephosphorylated Yap-1 signalling with verteporfin inhibited phorbol 12,13-dibutyrate/SB203580-mediated nestin expression and BrdU incorporation of neonatal cardiomyocytes. Nestin depletion with an AAV9 containing a shRNA directed against the intermediate filament protein significantly reduced the number of neonatal cardiomyocytes that re-entered the cell cycle. These findings demonstrate that Tbx5- and Gata4-dependent events negatively regulate nestin expression in cardiomyocytes during embryogenesis. By contrast, dephosphorylated Yap-1 acting via upregulation of the intermediate filament protein nestin plays a seminal role in the cell cycle re-entry of cardiomyocytes. Based on these data, an analogous role of Yap-1 may be prevalent in the heart of Tbx5+/- and Gata4+/- mice.
Collapse
Affiliation(s)
- Vanessa Hertig
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Adrianna Matos-Nieves
- Center for Cardiovascular Research and the Heart Center, Nationwide Children's Hospital, OH Department of Pediatrics, The Ohio State University, OH Department of Molecular Genetics, The Ohio State University, Columbus, Ohio
| | - Vidu Garg
- Center for Cardiovascular Research and the Heart Center, Nationwide Children's Hospital, OH Department of Pediatrics, The Ohio State University, OH Department of Molecular Genetics, The Ohio State University, Columbus, Ohio
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Maya Mamarbachi
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Laurie Caland
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada.,Department of Pharmacology & Physiology, Université de Montréal, Québec, Montréal, Canada
| | - Angelino Calderone
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada.,Department of Pharmacology & Physiology, Université de Montréal, Québec, Montréal, Canada
| |
Collapse
|
16
|
Saboor F, Reckmann AN, Tomczyk CUM, Peters DM, Weissmann N, Kaschtanow A, Schermuly RT, Michurina TV, Enikolopov G, Müller D, Mietens A, Middendorff R. Nestin-expressing vascular wall cells drive development of pulmonary hypertension. Eur Respir J 2016; 47:876-88. [PMID: 26699726 PMCID: PMC5796529 DOI: 10.1183/13993003.00574-2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 09/17/2015] [Indexed: 01/02/2023]
Abstract
Nestin, a well-known marker of neuronal stem cells, was recently suggested to characterise stem cell-like progenitors in non-neuronal structures during development and tissue repair. Integrating novel morphological approaches (CLARITY), we investigate whether nestin expression defines the proliferating cell population that essentially drives vascular remodelling during development of pulmonary hypertension.The role of nestin was investigated in lungs of nestin-GFP (green fluorescent protein) mice, models of pulmonary hypertension (rat: monocrotaline, SU5416/hypoxia; mouse: hypoxia), samples from pulmonary hypertension patients and human pulmonary vascular smooth muscle cells (VSMCs).Nestin was solely found in lung vasculature and localised to proliferating VSMCs, but not bronchial smooth muscle cells. Nestin was shown to affect cell number and was significantly enhanced in lungs early during development of pulmonary hypertension, correlating well with increased VSMC proliferation, expression of phosphorylated (activated) platelet-derived growth factor receptor β and downregulation of the smooth muscle cell differentiation marker calponin. At later time points when pulmonary hypertension became clinically evident, nestin expression and proliferation returned to control levels. Increase of nestin-positive VSMCs was also found in human pulmonary hypertension, both in vessel media and neointima.Nestin expression seems to be obligatory for VSMC proliferation, and specifies lung vascular wall cells that drive remodelling and (re-)generation. Our data promise novel diagnostic tools and therapeutic targets for pulmonary hypertension.
Collapse
Affiliation(s)
- Farhan Saboor
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany These authors contributed equally
| | - Ansgar N Reckmann
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany These authors contributed equally
| | - Claudia U M Tomczyk
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany These authors contributed equally
| | - Dorothea M Peters
- University of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Dept of Internal Medicine, Members of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| | - Norbert Weissmann
- University of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Dept of Internal Medicine, Members of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| | - Andre Kaschtanow
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Ralph T Schermuly
- University of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Dept of Internal Medicine, Members of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| | - Tatyana V Michurina
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA NBIC, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Grigori Enikolopov
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA NBIC, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Dieter Müller
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Andrea Mietens
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Ralf Middendorff
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
17
|
Vascular Wall-Resident Multipotent Stem Cells of Mesenchymal Nature within the Process of Vascular Remodeling: Cellular Basis, Clinical Relevance, and Implications for Stem Cell Therapy. Stem Cells Int 2016; 2016:1905846. [PMID: 26880936 PMCID: PMC4736960 DOI: 10.1155/2016/1905846] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/18/2015] [Indexed: 12/13/2022] Open
Abstract
Until some years ago, the bone marrow and the endothelial cell compartment lining the vessel lumen (subendothelial space) were thought to be the only sources providing vascular progenitor cells. Now, the vessel wall, in particular, the vascular adventitia, has been established as a niche for different types of stem and progenitor cells with the capacity to differentiate into both vascular and nonvascular cells. Herein, vascular wall-resident multipotent stem cells of mesenchymal nature (VW-MPSCs) have gained importance because of their large range of differentiation in combination with their distribution throughout the postnatal organism which is related to their existence in the adventitial niche, respectively. In general, mesenchymal stem cells, also designated as mesenchymal stromal cells (MSCs), contribute to the maintenance of organ integrity by their ability to replace defunct cells or secrete cytokines locally and thus support repair and healing processes of the affected tissues. This review will focus on the central role of VW-MPSCs within vascular reconstructing processes (vascular remodeling) which are absolute prerequisite to preserve the sensitive relationship between resilience and stability of the vessel wall. Further, a particular advantage for the therapeutic application of VW-MPSCs for improving vascular function or preventing vascular damage will be discussed.
Collapse
|
18
|
Chabot A, Hertig V, Boscher E, Nguyen QT, Boivin B, Chebli J, Bissonnette E, Villeneuve L, Brochiero E, Dupuis J, Calderone A. Endothelial and Epithelial Cell Transition to a Mesenchymal Phenotype Was Delineated by Nestin Expression. J Cell Physiol 2015; 231:1601-10. [DOI: 10.1002/jcp.25257] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/16/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Andréanne Chabot
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie Moléculaire et Intégrative; Université de Montréal; Montréal Quebéc Canada
| | - Vanessa Hertig
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie Moléculaire et Intégrative; Université de Montréal; Montréal Quebéc Canada
| | - Elena Boscher
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
| | - Quang Trinh Nguyen
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
| | - Benoît Boivin
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Biochimie et; Montréal Québec Canada
- Médecine; Université de Montréal; Montréal Québec Canada
| | | | - Elyse Bissonnette
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec; Département de Médicine; Université Laval; Québec Canada
| | - Louis Villeneuve
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
| | | | - Jocelyn Dupuis
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Médecine; Université de Montréal; Montréal Québec Canada
| | - Angelino Calderone
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie Moléculaire et Intégrative; Université de Montréal; Montréal Quebéc Canada
| |
Collapse
|
19
|
Tardif K, Hertig V, Duquette N, Villeneuve L, El-Hamamsy I, Tanguay JF, Calderone A. Nestin upregulation characterizes vascular remodeling secondary to hypertension in the rat. Am J Physiol Heart Circ Physiol 2015; 308:H1265-74. [PMID: 25770244 DOI: 10.1152/ajpheart.00804.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 02/03/2015] [Indexed: 11/22/2022]
Abstract
Proliferation and hypertrophy of vascular smooth muscle cells represent hallmark features of vessel remodeling secondary to hypertension. The intermediate filament protein nestin was recently identified in vascular smooth muscle cells and in other cell types directly participated in proliferation. The present study tested the hypothesis that vessel remodeling secondary to hypertension was characterized by nestin upregulation in vascular smooth muscle cells. Two weeks after suprarenal abdominal aorta constriction of adult male Sprague-Dawley rats, elevated mean arterial pressure increased the media area and thickness of the carotid artery and aorta and concomitantly upregulated nestin protein levels. In the normal adult rat carotid artery, nestin immunoreactivity was observed in a subpopulation of vascular smooth muscle cells, and the density significantly increased following suprarenal abdominal aorta constriction. Filamentous nestin was detected in cultured rat carotid artery- and aorta-derived vascular smooth muscle cells and an analogous paradigm observed in human aorta-derived vascular smooth muscle cells. ANG II and EGF treatment of vascular smooth muscle cells stimulated DNA and protein synthesis and increased nestin protein levels. Lentiviral short-hairpin RNA-mediated nestin depletion of carotid artery-derived vascular smooth muscle cells inhibited peptide growth factor-stimulated DNA synthesis, whereas protein synthesis remained intact. These data have demonstrated that vessel remodeling secondary to hypertension was characterized in part by nestin upregulation in vascular smooth muscle cells. The selective role of nestin in peptide growth factor-stimulated DNA synthesis has revealed that the proliferative and hypertrophic responses of vascular smooth muscle cells were mediated by divergent signaling events.
Collapse
Affiliation(s)
- Kim Tardif
- Program in Biomedical Sciences, Université de Montréal, Montréal, Québec, Canada; Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Vanessa Hertig
- Departement of Physiology, Université de Montréal, Montréal, Québec, Canada; and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Natacha Duquette
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Ismail El-Hamamsy
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Jean-François Tanguay
- Program in Biomedical Sciences, Université de Montréal, Montréal, Québec, Canada; Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Angelino Calderone
- Departement of Physiology, Université de Montréal, Montréal, Québec, Canada; and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
20
|
Chabot A, Meus MA, Naud P, Hertig V, Dupuis J, Villeneuve L, El Khoury N, Fiset C, Nattel S, Jasmin JF, Calderone A. Nestin is a Marker of Lung Remodeling Secondary to Myocardial Infarction and Type I Diabetes in the Rat. J Cell Physiol 2014; 230:170-9. [DOI: 10.1002/jcp.24696] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 06/05/2014] [Indexed: 01/25/2023]
Affiliation(s)
- Andréanne Chabot
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie; Université de Montréal; Montréal Québec Canada
| | - Marc-Andre Meus
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie; Université de Montréal; Montréal Québec Canada
| | - Patrice Naud
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
| | - Vanessa Hertig
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie; Université de Montréal; Montréal Québec Canada
| | - Jocelyn Dupuis
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
| | - Louis Villeneuve
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
| | - Nabel El Khoury
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie; Université de Montréal; Montréal Québec Canada
| | - Celine Fiset
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Faculté de Pharmacie; Université de Montréal; Montréal Québec Canada
| | - Stanley Nattel
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
| | - Jean-Francois Jasmin
- Department of Stem Cell Biology & Regenerative Medicine; Thomas Jefferson University; Philadelphia Pennsylvania
- Department of Pharmaceutical Sciences; University of the Sciences in Philadelphia; Philadelphia Pennsylvania
| | - Angelino Calderone
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie; Université de Montréal; Montréal Québec Canada
| |
Collapse
|
21
|
Yao Y, Norris EH, Strickland S. The cellular origin of laminin determines its role in blood pressure regulation. Cell Mol Life Sci 2014; 72:999-1008. [PMID: 25216704 DOI: 10.1007/s00018-014-1732-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 08/11/2014] [Accepted: 09/08/2014] [Indexed: 11/26/2022]
Abstract
Laminin of different cellular sources has distinct functions. In addition to vascular smooth muscle cells (SMCs), aorta also contains a small population of nestin(+) cells, whose function remains unknown. This study investigates the role of SMC- and nestin(+) cell-derived laminin in blood pressure (BP) regulation and SMC contractibility. Using mice with laminin deficiency in SMCs (SKO) or nestin(+) cells (NKO), we examined laminin-dependent changes in BP. Contractile protein expression was reduced in SKO but not NKO mice, consistent with their, respectively, low and normal baseline BP measurements. At the ultrastructural level, SKO SMCs maintained the contractile phenotype with reduced elasticity, whereas NKO SMCs switched to the synthetic phenotype and showed degeneration. Additionally, angiotensin II (Ang II) significantly increased BP in SKO but not NKO mice. It also enhanced contractile proteins to the same levels and induced SMC degeneration in both knockout mice. These data suggest that SMC laminin regulates BP via modulating contractile protein expression, whereas nestin(+) cell-derived laminin contributes to SMC phenotypic switch.
Collapse
Affiliation(s)
- Yao Yao
- Laboratory of Neurobiology and Genetics, The Rockefeller University, 1230 York Ave, Box 169, New York, NY, 10065, USA
| | | | | |
Collapse
|
22
|
Tardif K, Hertig V, Dumais C, Villeneuve L, Perrault L, Tanguay JF, Calderone A. Nestin downregulation in rat vascular smooth muscle cells represents an early marker of vascular disease in experimental type I diabetes. Cardiovasc Diabetol 2014; 13:119. [PMID: 25139503 PMCID: PMC4143548 DOI: 10.1186/s12933-014-0119-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/23/2014] [Indexed: 01/28/2023] Open
Abstract
Background Nestin was reported to directly contribute to cell proliferation and the intermediate filament protein was detected in vascular smooth muscle cells. In experimental type I diabetes, nestin downregulation in the heart was identified as an incipient pathophysiological event. The following study tested the hypothesis that dysregulation of nestin expression in vascular smooth muscle cells represented an early event of vascular disease in experimental type I diabetes. Methods/Results In the carotid artery and aorta of adult male Sprague-Dawley rats, a subpopulation of vascular smooth muscle cells co-expressed nestin and was actively involved in the cell cycle as reflected by the co-staining of nuclear phosphohistone-3. The infection of aortic vascular smooth muscle cells with a lentivirus containing a shRNAmir directed against nestin significantly reduced protein expression and concomitantly attenuated basal DNA synthesis. Two weeks following injection of adult male Sprague-Dawley rats with streptozotocin, the endothelial response of aortic rings to acetylcholine, vascular morphology and the total density of vascular smooth muscle cells in the vasculature of type I diabetic rats were similar to normal rats. By contrast, nestin protein levels and the density of nestin(+)/phosphohistone-3(+)-vascular smooth muscle cells were significantly reduced in type I diabetic rats. The in vivo observations were recapitulated in vitro as exposure of vascular smooth muscle cells to 30 mM D-glucose inhibited DNA synthesis and concomitantly reduced nestin protein expression. Conclusions Hyperglycaemia-mediated nestin downregulation and the concomitant reduction of cycling vascular smooth muscle cells represent early markers of vascular disease in experimental type I diabetes. Electronic supplementary material The online version of this article (doi:10.1186/s12933-014-0119-6) contains supplementary material, which is available to authorized users.
Collapse
|
23
|
Astrocyte activation is suppressed in both normal and injured brain by FGF signaling. Proc Natl Acad Sci U S A 2014; 111:E2987-95. [PMID: 25002516 DOI: 10.1073/pnas.1320401111] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the brain, astrocytes are multifunctional cells that react to insults and contain damage. However, excessive or sustained reactive astrocytes can be deleterious to functional recovery or contribute to chronic inflammation and neuronal dysfunction. Therefore, astrocyte activation in response to damage is likely to be tightly regulated. Although factors that activate astrocytes have been identified, whether factors also exist that maintain astrocytes as nonreactive or reestablish their nonreactive state after containing damage remains unclear. By using loss- and gain-of-function genetic approaches, we show that, in the unperturbed adult neocortex, FGF signaling is required in astrocytes to maintain their nonreactive state. Similarly, after injury, FGF signaling delays the response of astrocytes and accelerates their deactivation. In addition, disrupting astrocytic FGF receptors results in reduced scar size without affecting neuronal survival. Overall, this study reveals that the activation of astrocytes in the normal and injured neocortex is not only regulated by proinflammatory factors, but also by factors such as FGFs that suppress activation, providing alternative therapeutic targets.
Collapse
|
24
|
Adult vascular smooth muscle cells in culture express neural stem cell markers typical of resident multipotent vascular stem cells. Cell Tissue Res 2014; 358:203-16. [PMID: 24992927 DOI: 10.1007/s00441-014-1937-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 05/28/2014] [Indexed: 10/25/2022]
Abstract
Differentiation of resident multipotent vascular stem cells (MVSCs) or de-differentiation of vascular smooth muscle cells (vSMCs) might be responsible for the SMC phenotype that plays a major role in vascular diseases such as arteriosclerosis and restenosis. We examined vSMCs from three different species (rat, murine and bovine) to establish whether they exhibit neural stem cell characteristics typical of MVSCs. We determined their SMC differentiation, neural stem cell marker expression and multipotency following induction in vitro by using immunocytochemistry, confocal microscopy, fluorescence-activated cell sorting analysis and quantitative real-time polymerase chain reaction. MVSCs isolated from rat aortic explants, enzymatically dispersed rat SMCs and rat bone-marrow-derived mesenchymal stem cells served as controls. Murine carotid artery lysates and primary rat aortic vSMCs were both myosin-heavy-chain-positive but weakly expressed the neural crest stem cell marker, Sox10. Each vSMC line examined expressed SMC differentiation markers (smooth muscle α-actin, myosin heavy chain and calponin), neural crest stem cell markers (Sox10(+), Sox17(+)) and a glia marker (S100β(+)). Serum deprivation significantly increased calponin and myosin heavy chain expression and decreased stem cell marker expression, when compared with serum-rich conditions. vSMCs did not differentiate to adipocytes or osteoblasts following adipogenic or osteogenic inductive stimulation, respectively, or respond to transforming growth factor-β1 or Notch following γ-secretase inhibition. Thus, vascular SMCs in culture express neural stem cell markers typical of MVSCs, concomitant with SMC differentiation markers, but do not retain their multipotency. The ultimate origin of these cells might have important implications for their use in investigations of vascular proliferative disease in vitro.
Collapse
|
25
|
EGF-induced adipose tissue mesothelial cells undergo functional vascular smooth muscle differentiation. Cell Death Dis 2014; 5:e1304. [PMID: 24967966 PMCID: PMC4611741 DOI: 10.1038/cddis.2014.271] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/01/2014] [Accepted: 05/20/2014] [Indexed: 12/12/2022]
Abstract
Recent studies suggested that the post-natal mesothelium retain differentiative potential of the embryonic mesothelium, which generates fibroblasts and vascular smooth muscle cells (VSMCs), in developing coelomic organs via epithelial-to-mesenchymal transition (EMT). Whether adult mesothelial cells (MCs) are able to give rise to functional VSMCs in vitro and which are the factors and mechanisms directing this process remain largely unknown. Here, we isolated adipose tissue MCs (ATMCs) from adult mice, and demonstrated that ATMCs cultured in a serum-containing media supplemented with epidermal growth factor (EGF) efficiently increased both their proliferation and EMT above levels found in only serum-containing media cultures. EGF-induced ATMCs gained phosphorylation of the EGF receptor and activated simultaneously ILK/Erk1/2, PI3K/Akt and Smad2/3-dependent pathways. Sequential subculture onto collagen-I surface efficiently improved their vasculogenic EMT towards cells featuring VSMCs (α-SMA, calponin, caldesmon, SM22α, desmin, SM-MHC, smoothelin-B and PDGFR-β) that could actively contract in response to receptor and non-receptor-mediated vasoactive agonists. Overall, our results indentify EGF signalling as a robust vasculogenic inductive pathway for ATMCs, leading to their transdifferentiation into functional VSMC-like cells.
Collapse
|
26
|
Klein D, Meissner N, Kleff V, Jastrow H, Yamaguchi M, Ergün S, Jendrossek V. Nestin(+) tissue-resident multipotent stem cells contribute to tumor progression by differentiating into pericytes and smooth muscle cells resulting in blood vessel remodeling. Front Oncol 2014; 4:169. [PMID: 25019063 PMCID: PMC4072089 DOI: 10.3389/fonc.2014.00169] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 06/11/2014] [Indexed: 12/14/2022] Open
Abstract
Tumor vessels with resistance to anti-angiogenic therapy are characterized by the normalization of the vascular structures through integration of mature pericytes and smooth muscle cells (SMC) into the vessel wall, a process termed vessel stabilization. Unfortunately, stabilization-associated vascular remodeling can result in reduced sensitivity to subsequent anti-angiogenic therapy. We show here that blockade of VEGF by bevacizumab induces stabilization of angiogenic tumor blood vessels in human tumor specimen by recruiting Nestin-positive cells, whereas mature vessels down-regulated Nestin-expression. Using xenograft tumors growing on bone-marrow (BM) chimera of C57Bl/6 wildtype and Nestin-GFP transgenic mice, we show for first time that Nestin(+) cells inducing the maturation of tumor vessels do not originate from the BM but presumably reside within the adventitia of adult blood vessels. Complementary ex vivo experiments using explants of murine aortas revealed that Nestin(+) multipotent stem cells (MPSCs) are mobilized from their niche and differentiated into pericytes and SMC through the influence of tumor-cell-secreted factors. We conclude that tissue-resident Nestin(+) cells are more relevant than BM-derived cells for vessel stabilization and therefore have to be considered in future strategies for anti-angiogenic therapy. The identification of proteins mediating recruitment or differentiation of local Nestin(+) cells with potential stem cell character to angiogenic blood vessels may allow the definition of new therapeutic targets to reduce tumor resistance against anti-angiogenic drugs.
Collapse
Affiliation(s)
- Diana Klein
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen , Essen , Germany
| | - Nicole Meissner
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen , Essen , Germany
| | - Veronika Kleff
- Institute of Anatomy, University Hospital, University of Duisburg-Essen , Essen , Germany
| | - Holger Jastrow
- Institute of Anatomy, University Hospital, University of Duisburg-Essen , Essen , Germany
| | - Masahiro Yamaguchi
- Department of Physiology, Graduate School of Medicine, University of Tokyo , Tokyo , Japan
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, University of Würzburg , Würzburg , Germany
| | - Verena Jendrossek
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen , Essen , Germany
| |
Collapse
|
27
|
Vascular and parenchymal lesions along with enhanced neurogenesis characterize the brain of asymptomatic stroke-prone spontaneous hypertensive rats. J Hypertens 2014; 31:1618-28. [PMID: 23666422 DOI: 10.1097/hjh.0b013e3283619d7f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND OBJECTIVES Spontaneously hypertensive stroke-prone rats (SHRSPs) develop hypertension, cerebrovascular abnormalities and a stroke phenotype in association with higher levels of proteinuria. Here, we focus on cerebral abnormalities preceding lesions detectable by MRI. METHODS Longitudinal assessment of brain histology was performed in salt-loaded male SHRSPs (n = 26) and Wistar-Kyoto (WKY) normotensive control animals (n = 27). Groups of rats were sacrificed at different time points: Time 0, before the salt diet administration; Time 1, when proteinuria achieved 40 mg/day; Time 2, when proteinuria exceeded 100 mg/day. RESULTS At Time 0, no brain lesions were observed. At Time 1, changes of the cortical penetrating arteries, vasogenic oedema, lacunae and focal cell loss appeared in SHRSPs and worsened at Time 2, although no lesions were yet detected by MRI. Staining for proliferation markers revealed a significant boost of cellular mitosis in the subventricular zone (SVZ) of SHRSPs. Moreover, we observed higher immunopositivity for nestin, glial fibrillary acidic protein and doublecortin (markers for neural stem cells, astrocytes and immature neurons, respectively). At Time 2, apoptotic caspase-3 as well as 4-hydroxynonenal-positive neurons were associated to decreased nestin and doublecortin staining. High expression levels of glial fibrillary acidic protein were maintained in the SVZ. No comparative alterations and SVZ activation were recorded in WKYs. CONCLUSION Appearance of vascular changes in SHRSPs, before any MRI-detectable brain lesion, is coupled to active neural proliferation in the SVZ. With disease progression, only newborn astrocytes can survive, likely because of the neurotoxicity triggered by brain oedema and oxidative stress.
Collapse
|
28
|
Chang KW, Huang YL, Wong ZR, Su PH, Huang BM, Ju TK, Yang HY. Fibroblast growth factor-2 up-regulates the expression of nestin through the Ras-Raf-ERK-Sp1 signaling axis in C6 glioma cells. Biochem Biophys Res Commun 2013; 434:854-60. [PMID: 23611784 DOI: 10.1016/j.bbrc.2013.04.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 04/04/2013] [Indexed: 11/30/2022]
Abstract
Nestin is a 240-kDa intermediate filament protein expressed mainly in neural and myogenic stem cells. Although a substantial number of studies have focused on the expression of nestin during development of the central nervous system, little is known about the factors that induce and regulate its expression. Fibroblast growth factor-2 (FGF-2) is an effective mitogen and stimulates the proliferation and differentiation of a subset of nestin-expressing cells, including neural progenitor cells, glial precursor cells, and smooth muscle cells. To assess whether FGF-2 is a potent factor that induces the expression of nestin, C6 glioma cells were used. The results showed that nestin expression was up-regulated by FGF-2 via de novo RNA and protein synthesis. Our RT-PCR results showed that C6 glioma cells express FGFR1/3, and FGFRs is required for FGF-2-induced nestin expression. Further signaling analysis also revealed that FGF-2-induced nestin expression is mediated through FGFR-MAPK-ERK signaling axis and the transcriptional factor Sp1. These findings provide new insight into the regulation of nestin in glial system and enable the further studies on the function of nestin in glial cells.
Collapse
Affiliation(s)
- Kai-Wei Chang
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 106, Taiwan
| | | | | | | | | | | | | |
Collapse
|
29
|
Induction of vascular endothelial growth factor receptor-3 expression in perivascular cells of the ischemic core following focal cerebral ischemia in rats. Acta Histochem 2013; 115:170-7. [PMID: 22771250 DOI: 10.1016/j.acthis.2012.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 06/16/2012] [Accepted: 06/18/2012] [Indexed: 01/02/2023]
Abstract
Vascular endothelial growth factor receptor (VEGFR)-3, a receptor for VEGF-C and VEGF-D, has recently been reported to be induced within vessel-like structures in the ischemic brain. The purpose of the present study was to characterize and define further the cellular phenotypes of vascular-associated cells that manifest induced VEGFR-3 expression in a rat model of ischemic stroke. Vessel-associated cells expressing VEGFR-3 were found to be perivascular astrocytes in the peri-infarct region, whereas in the ischemic core, where astrocytes had virtually disappeared, induction of VEGFR-3 mRNA and protein was still prominent in vascular structures 3-7 days after reperfusion. VEGFR-3 and nestin expression were colocated in almost all cells associated with the vasculature in the ischemic core, and most (~82%) of the VEGFR-3/nestin double-labeled cells were proliferative. A subpopulation of these VEGFR-3-expressing cells appeared to be included in two immunophenotypically distinct perivascular cells: NG2-positive pericytes and ED2- or OX6-perivascular macrophages. However, most of these cells did not show markers for vasculature-associated cell types such as endothelial cells, microglia/macrophages, and smooth muscle cells. Thus, our data indicated that vasculature-associated VEGFR-3-expressing cells in the ischemic core may represent a heterogeneous population of cells with functional diversity, rather than a uniform cell type.
Collapse
|
30
|
Lachaud CC, Pezzolla D, Domínguez-Rodríguez A, Smani T, Soria B, Hmadcha A. Functional vascular smooth muscle-like cells derived from adult mouse uterine mesothelial cells. PLoS One 2013; 8:e55181. [PMID: 23405120 PMCID: PMC3566215 DOI: 10.1371/journal.pone.0055181] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/19/2012] [Indexed: 12/13/2022] Open
Abstract
In mammalian visceral organs, vascular smooth muscle cells (VSMCs) originate from an epithelial-to-mesenchymal transition (EMT) of embryonic mesothelial cells (MCs). The ability of adult MCs to recapitulate EMT and to acquire smooth muscle (SM) markers upon provasculogenic culture suggested they might retain embryonic vasculogenic differentiation potential. However, it remains unknown whether adult MCs-derived SM-like cells may acquire specific vascular SM lineage markers and the functionality of differentiated contractile VSMCs. Here, we describe how a gentle trypsinization of adult mouse uterine cords could selectively detach their outermost uterine mesothelial layer cells. As other MCs; uterine MCs (UtMCs) uniformly expressed the epithelial markers β-catenin, ZO-1, E-cadherin, CD54, CD29, and CK18. When cultured in a modified SM differentiation media (SMDM) UtMCs initiated a loss of epithelial characteristics and gained markers expression of EMT (Twist, Snail, and Slug), stem and progenitor (Nanog, Sox2, C-kit, Gata-4, Isl-1, and nestin), SM (α-SMA, calponin, caldesmon, SM22α, desmin, SM-MHC, and smoothelin-B) and cardiac (BMP2, BMP4, ACTC1, sACTN, cTnI, cTnT, ANF, Cx43, and MLC2a). UtMCs repeatedly subcultured in SMDM acquired differentiated VSM-like characteristics and expressed smoothelin-B in the typical stress-fiber pattern expression of contractile VSMCs. Relevantly, UtMCs-derived VSM-like cells could generate "mechanical force" to compact collagen lattices and displayed in diverse degree voltage (K(+)) and receptor (endothelin-1, oxytocin, norepinephrine, carbachol and vasopressin)-induced [Ca(2+)](i) rises and contraction. Thus, we show for the first time that UtMCs could recapitulate in vitro differentiative events of early cardiovascular differentiation and transdifferentiate in cells exhibiting molecular and functional characteristics of VSMCs.
Collapse
Affiliation(s)
| | - Daniela Pezzolla
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Sevilla, Spain
| | | | - Tarik Smani
- Instituto de Biomedicina de Sevilla/Fisiopatología Cardiovascular, Sevilla, Spain
| | - Bernat Soria
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Sevilla, Spain
- CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Barcelona, Spain
| | - Abdelkrim Hmadcha
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Sevilla, Spain
- CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Barcelona, Spain
| |
Collapse
|
31
|
Su PH, Chen CC, Chang YF, Wong ZR, Chang KW, Huang BM, Yang HY. Identification and cytoprotective function of a novel nestin isoform, Nes-S, in dorsal root ganglia neurons. J Biol Chem 2013; 288:8391-8404. [PMID: 23319587 DOI: 10.1074/jbc.m112.408179] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In this study, the first nestin isoform, Nes-S, was identified in neurons of dorsal root ganglia (DRG) of adult rats. Nes-S cannot form filaments by itself in cytoplasmic intermediate filament-free SW13 cells. Instead, it co-assembles into filaments with vimentin when transfected into vimentin(+) SW13 cells, and with peripherin and neurofilament proteins when transfected into N2a cells. In primary DRG neurons, endogenous Nes-S co-assembles with peripherin and neurofilament proteins. The expression of Nes-S first appears in DRG at postnatal day 5 and persists to adulthood. Among the adult tissues we examined, the expression of Nes-S is restricted to the sensory and motor neurons. Finally, exogenous Nes-S enhances viability when transfected into N2a cells, and knockdown of endogenous Nes-S impairs the survival of DRG neurons in primary cultures. Taken together, Nes-S is a new neuronal intermediate filament protein that exerts a cytoprotective function in mature sensory and motor neurons.
Collapse
Affiliation(s)
- Peng-Han Su
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 10617, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Ya-Fan Chang
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 10617, Taiwan
| | - Zong-Ruei Wong
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 10617, Taiwan
| | - Kai-Wei Chang
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 10617, Taiwan
| | - Bu-Miin Huang
- Department of Cell Biology and Anatomy, National Cheng-Kung University, Tainan 701, Taiwan
| | - Hsi-Yuan Yang
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
32
|
Characterization of nestin expression and vessel association in the ischemic core following focal cerebral ischemia in rats. Cell Tissue Res 2012; 351:383-95. [DOI: 10.1007/s00441-012-1538-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/23/2012] [Indexed: 12/23/2022]
|
33
|
Béguin PC, Gosselin H, Mamarbachi M, Calderone A. Nestin expression is lost in ventricular fibroblasts during postnatal development of the rat heart and re-expressed in scar myofibroblasts. J Cell Physiol 2012; 227:813-20. [PMID: 21503881 DOI: 10.1002/jcp.22794] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Studies have reported that the intermediate filament protein nestin was expressed in various non-stem/progenitor cells during development, downregulated during postnatal growth and re-expressed following injury. The present study tested the hypothesis that an analogous paradigm was prevalent for ventricular fibroblasts. In the neonatal rat heart, nestin protein levels were significantly higher than the adult heart and the isolation of cardiac cells revealed a selective expression in ventricular fibroblasts. In adult ventricular fibroblasts, nestin protein expression was markedly lower compared to neonatal ventricular fibroblasts. Following ischemic damage to the rat heart, nestin staining was detected in a subpopulation of scar myofibroblasts (37%) and the percentage of immunoreactive cells was greater than adult ventricular fibroblasts (7%) but significantly lower than neonatal ventricular fibroblasts (86%). Moreover, dissimilar rates of (3)H-thymidine uptake were observed among the fibroblast populations and may be related in part to the disparate percentage of nestin(+) cells. To assess the role of nestin in DNA synthesis, neonatal ventricular fibroblasts were infected with a lentivirus containing a shRNAmir directed against the intermediate filament protein. The partial depletion of nestin expression in neonatal ventricular fibroblasts significantly reduced basal DNA synthesis, in the absence of an apoptotic response. Thus, postnatal development of the rat heart was associated with a selective loss of nestin expression in ventricular fibroblasts and subsequent induction in a subpopulation of myofibroblasts following ischemic injury. The re-expression of nestin in scar myofibroblasts may represent an adaptive response to enhance their proliferative rate and accelerate the healing process.
Collapse
Affiliation(s)
- Pauline C Béguin
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
34
|
Abstract
Protocols are presented describing a unique in vitro injury model and how to culture and mature mouse, rat, and human astrocytes for its use. This injury model produces widespread injury and astrocyte reactivity that enable quantitative measurements of morphological, biochemical, and functional changes in rodent and human reactive astrocytes. To investigate structural and molecular mechanisms of reactivity in vitro, cultured astrocytes need to be purified and then in vitro "matured" to reach a highly differentiated state. This is achieved by culturing astrocytes on deformable collagen-coated membranes in the presence of adult-derived horse serum (HS), followed by its stepwise withdrawal. These in vitro matured, process-bearing, quiescent astrocytes are then subjected to mechanical stretch injury by an abrupt pressure pulse from a pressure control device that briefly deforms the culture well bottom. This inflicts a measured reproducible, widespread strain that induces reactivity and injury in rodent and human astrocytes. Cross-species comparisons are possible because mouse, rat, and human astrocytes are grown using essentially the same in vitro treatment regimen. Human astrocytes from fetal cerebral cortex are compared to those derived from cortical biopsies of epilepsy patients (ages 1-12 years old), with regard to growth, purity, and differentiation. This opens a unique opportunity for future studies on glial biology, maturation, and pathology of human astrocytes. Prototypical astrocyte proteins including GFAP, S100, aquaporin4, glutamate transporters, and tenascin are expressed in mouse, rat, and human in vitro matured astrocyte. Upon pressure-stretching, rodent and human astrocytes undergo dynamic morphological, gene expression, and protein changes that are characteristic for trauma-induced reactive astrogliosis.
Collapse
Affiliation(s)
- Ina-Beate Wanner
- Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
35
|
Abstract
Scar formation following an ischemic insult to the heart is referred to as reparative fibrosis and represents an essential physiological response to heal the damaged myocardium. The biological events of reparative fibrosis include inflammation, the deposition of collagen by myofibroblasts, sympathetic innervation, and angiogenesis. Several studies have further reported that scar formation was associated with the recruitment of neural crest-derived cardiac resident nestin(+) cells that display characteristics consistent with a neural progenitor/stem cell phenotype. During the reparative fibrotic response, these nestin(+) cells participate in neural remodeling and represent a novel cellular substrate of angiogenesis. In addition, a subpopulation of nestin(+) cells identified in the normal heart expressed cardiac progenitor transcriptional factors and may directly contribute to myocardial regeneration following ischemic damage. Nestin protein was also detected in endothelial cells of newly formed blood vessels in the scar and may represent a marker of revascularization. Lastly, nestin was induced in a subpopulation of smooth muscle α-actin(+) scar-derived myofibroblasts, and the expression of the intermediate filament protein may provide a proliferative advantage. Collectively, these data demonstrate that diverse populations of nestin(+) cells participate in cardiac wound healing.
Collapse
|