1
|
Rasile M, Lauranzano E, Faggiani E, Ravanelli MM, Colombo FS, Mirabella F, Corradini I, Malosio ML, Borreca A, Focchi E, Pozzi D, Giorgino T, Barajon I, Matteoli M. Maternal immune activation leads to defective brain-blood vessels and intracerebral hemorrhages in male offspring. EMBO J 2022; 41:e111192. [PMID: 36314682 PMCID: PMC9713716 DOI: 10.15252/embj.2022111192] [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: 03/16/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 12/04/2022] Open
Abstract
Intracerebral hemorrhages are recognized risk factors for neurodevelopmental disorders and represent early biomarkers for cognitive dysfunction and mental disability, but the pathways leading to their occurrence are not well defined. We report that a single intrauterine exposure of the immunostimulant Poly I:C to pregnant mice at gestational day 9, which models a prenatal viral infection and the consequent maternal immune activation, induces the defective formation of brain vessels and causes intracerebral hemorrhagic events, specifically in male offspring. We demonstrate that maternal immune activation promotes the production of the TGF-β1 active form and the consequent enhancement of pSMAD1-5 in males' brain endothelial cells. TGF-β1, in combination with IL-1β, reduces the endothelial expression of CD146 and claudin-5, alters the endothelium-pericyte interplay resulting in low pericyte coverage, and increases hemorrhagic events in the adult offspring. By showing that exposure to Poly I:C at the beginning of fetal cerebral angiogenesis results in sex-specific alterations of brain vessels, we provide a mechanistic framework for the association between intragravidic infections and anomalies of the neural vasculature, which may contribute to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Marco Rasile
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | | | - Elisa Faggiani
- IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | - Margherita M Ravanelli
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | | | - Filippo Mirabella
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | - Irene Corradini
- IRCCS Humanitas Clinical and Research CenterRozzanoItaly,Institute of Neuroscience (IN‐CNR)National Research Council of ItalyMilanItaly
| | - Maria L Malosio
- IRCCS Humanitas Clinical and Research CenterRozzanoItaly,Institute of Neuroscience (IN‐CNR)National Research Council of ItalyMilanItaly
| | - Antonella Borreca
- IRCCS Humanitas Clinical and Research CenterRozzanoItaly,Institute of Neuroscience (IN‐CNR)National Research Council of ItalyMilanItaly
| | - Elisa Focchi
- Institute of Neuroscience (IN‐CNR)National Research Council of ItalyMilanItaly
| | - Davide Pozzi
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | - Toni Giorgino
- Institute of Biophysics (IBF‐CNR)National Research Council of ItalyMilanItaly
| | - Isabella Barajon
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,IRCCS Humanitas Clinical and Research CenterRozzanoItaly
| | - Michela Matteoli
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly,Institute of Neuroscience (IN‐CNR)National Research Council of ItalyMilanItaly
| |
Collapse
|
2
|
He X, Wu DF, Ji J, Ling WP, Chen XL, Chen YX. Ultrasound microbubble-carried PNA targeting to c-myc mRNA inhibits the proliferation of rabbit iliac arterious smooth muscle cells and intimal hyperplasia. Drug Deliv 2015; 23:2482-2487. [PMID: 25726989 DOI: 10.3109/10717544.2015.1014947] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To elucidate the transfected effect of albumin ultrasound microbubbles carrying peptide nucleic acids (PNAs) against c-myc gene to the vascular walls and their effect on the intimal proliferation induced by vascular denudation. METHODS A rabbit iliac artery intimal proliferation model was constructed and PNA against c-myc mRNA was designed and synthesized and was added to albumin solution before ultrasound microbubbles were prepared and encapsulated in matrix of albumin. The ultrasound microbubbles carrying PNA were transfected to intima under ultrasound exposure. The transfected effect was identified by a histochemical method and the expression of c-myc was detected by in situ hybridization. The proliferation of intimal smooth muscle cells was estimated by the expression of proliferative cell nuclear antigen (PCNA) of them. The intimal area and thickness were judged morphologically for intimal hyperplasia. RESULTS The ultrasound microbubbles with PNA were successfully prepared and c-myc PNA was transfected to vascular intimal cells. The expression of c-myc and PCNA by intimal vascular smooth muscle cells (vSMCs) was inhibited significantly and the intimal thickness and area were reduced remarkably. CONCLUSION Transfection of c-myc PNA could inhibit proliferartion of vSMCs and intima in the rabbit iliac artery intimal proliferation model and the targeted transfection of albumin ultrasound microbubbles carrying PNA offers a feasible way to facilitate its access to specific cells in vivo and produce bioavailability.
Collapse
Affiliation(s)
- Xia He
- a The Department of Pathology , Shenzhen Sun Yet-Sen Cardiovascular Hospital , Shenzhen , PR China and
| | - Da-Fang Wu
- b Department of Endocrinology , 451 Hospital of PLA , Xi'an, Shanxi , PR China
| | - Jun Ji
- a The Department of Pathology , Shenzhen Sun Yet-Sen Cardiovascular Hospital , Shenzhen , PR China and
| | - Wen-Ping Ling
- a The Department of Pathology , Shenzhen Sun Yet-Sen Cardiovascular Hospital , Shenzhen , PR China and
| | - Xiao-Ling Chen
- a The Department of Pathology , Shenzhen Sun Yet-Sen Cardiovascular Hospital , Shenzhen , PR China and
| | - Yue-Xuan Chen
- a The Department of Pathology , Shenzhen Sun Yet-Sen Cardiovascular Hospital , Shenzhen , PR China and
| |
Collapse
|
3
|
Anderson JL, Smith SC, Taylor RL. The pigeon (Columba livia) model of spontaneous atherosclerosis. Poult Sci 2014; 93:2691-9. [PMID: 25214557 DOI: 10.3382/ps.2014-04280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Multiple animal models have been employed to study human atherosclerosis, the principal cause of mortality in the United States. Each model has individual advantages related to specific pathologies. Initiation, the earliest disease phase, is best modeled by the White Carneau (WC-As) pigeon. Atherosclerosis develops spontaneously in the WC-As without either external manipulation or known risk factors. Furthermore, susceptibility is caused by a single gene defect inherited in an autosomal recessive manner. The Show Racer (SR-Ar) pigeon is resistant to atherosclerosis. Breed differences in the biochemistry and metabolism of celiac foci cells have been described. For example, WC-As have lower oxidative metabolism but higher amounts of chondroitin-6-sulfate and nonesterified fatty acids compared with SR-Ar. Gene expression in aortic smooth muscle cells was compared between breeds using representational difference analysis and microarray analysis. Energy metabolism and cellular phenotype were the chief gene expression differences. Glycolysis and synthetic cell types were related to the WC-As but oxidative metabolism and contractile cell types were related to the SR-Ar. Rosiglitazone, a PPARγ agonist, blocked RNA binding motif (RBMS1) expression in WC-As cells. The drug may act through the c-myc oncogene as RBMS1 is a c-myc target. Proteomic tests of aortic smooth muscle cells supported greater glycosylation in the WC-As and a transforming growth factor β effect in SR-Ar. Unoxidized fatty acids build up in WC-As cells because of their metabolic deficiency, ultimately preventing the contractile phenotype in these cells. The single gene responsible for the disease is likely regulatory in nature.
Collapse
Affiliation(s)
- J L Anderson
- Department of Animal and Nutritional Sciences, University of New Hampshire, Durham 03824
| | - S C Smith
- Department of Animal and Nutritional Sciences, University of New Hampshire, Durham 03824
| | - R L Taylor
- Department of Animal and Nutritional Sciences, University of New Hampshire, Durham 03824
| |
Collapse
|
4
|
Xu C, Lu C, Hua L, Jin H, Yin L, Chen S, Qian R. Rhythm changes of clock genes, apoptosis-related genes and atherosclerosis-related genes in apolipoprotein E knockout mice. Can J Cardiol 2009; 25:473-9. [PMID: 19668782 DOI: 10.1016/s0828-282x(09)70122-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Acute myocardial infarction and stroke occur more frequently in the morning, suggesting a role of the circadian clock in these main causes of death, secondary to atherosclerosis. OBJECTIVES To investigate the expression of clock genes, apoptosis-related genes and atherosclerosis-related genes in the process of atherosclerosis. METHODS Apolipoprotein E knockout (ApoE-/-) mice were used to establish animal models of early and advanced atherosclerosis. Real-time polymerase chain reaction, Western blotting and microarray assays were used to detect the expression of clock genes, apoptosis-related genes and atherosclerosis-related genes. RESULTS Clock genes in ApoE-/- and C57BL/6J mouse hearts exhibited daily oscillations at the messenger RNA level. However, the expression level and rhythm between ApoE-/- and C57BL/6J mice were significantly different. Moreover, the changes became more significant as atherosclerosis developed. c-Myc and p53 genes exhibited circadian expression in C57BL/6J mice at messenger RNA and protein levels. However, the rhythm in ApoE-/- mice disappeared completely. Bcl-2 and Bax did not show daily rhythm in either strain of mouse. Aside from apoptosis-related genes, several atherosclerosis-related genes expressed time-dependent behaviour in C57BL/6J mice but not in ApoE-/- mice. CONCLUSIONS Rhythm changes of clock genes, apoptosis-related genes and atherosclerosis-related genes may play important roles in atherosclerosis and its complications.
Collapse
Affiliation(s)
- Chen Xu
- Department of Physiology and Pathophysiology, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
5
|
Kona S, Chellamuthu P, Xu H, Hills SR, Nguyen KT. Effects of cyclic strain and growth factors on vascular smooth muscle cell responses. Open Biomed Eng J 2009; 3:28-38. [PMID: 19812708 PMCID: PMC2757671 DOI: 10.2174/1874120700903010028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Revised: 07/06/2009] [Accepted: 07/09/2009] [Indexed: 01/09/2023] Open
Abstract
Under physiological and pathological conditions, vascular smooth muscle cells (SMC) are exposed to different biochemical factors and biomechanical forces. Previous studies pertaining to SMC responses have not investigated the effects of both factors on SMCs. Thus, in our research we investigated the combined effects of growth factors like Bfgf (basic fibroblast growth factor), TGF-β (transforming growth factor β) and PDGF (platelet-derived growth factor) along with physiological cyclic strain on SMC responses. Physiological cyclic strain (10% strain) significantly reduced SMC proliferation compared to static controls while addition of growth factors bFGF, TGF-β or PDGF-AB had a positive influence on SMC growth compared to strain alone. Microarray analysis of SMCs exposed to these growth factors and cyclic strain showed that several bioactive genes (vascular endothelial growth factor, epidermal growth factor receptor, etc.) were altered upon exposure. Further work involving biochemical and pathological cyclic strain stimulation will help us better understand the role of cyclic strain and growth factors in vascular functions and development of vascular disorders.
Collapse
Affiliation(s)
- Soujanya Kona
- Department of Bioengineering, University of Texas at Arlington and University of Texas Southwestern Medical Center at Dallas, USA
| | | | | | | | | |
Collapse
|
6
|
Khanna AK. Enhanced susceptibility of cyclin kinase inhibitor p21 knockout mice to high fat diet induced atherosclerosis. J Biomed Sci 2009; 16:66. [PMID: 19604372 PMCID: PMC2720941 DOI: 10.1186/1423-0127-16-66] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Accepted: 07/15/2009] [Indexed: 11/10/2022] Open
Abstract
Cyclin kinase inhibitor p21 is one of the most potent inhibitors of aortic smooth muscle cell proliferation, a key mediator of atherosclerosis. This study tests if p2l deficiency will result in severe atherosclerosis in a mouse model. p21-/- and strain matched wild type mice were fed with high fat diet for 21 weeks. Analysis for biochemical parameters (cholesterol, triglycerides) in serum and mRNA expression of CD36, HO-1, TGF-beta, IFN-gamma, TNF-alpha, PPAR-gamma and NADPH oxidase components (p22phox, NOX-1 and Rac-1) was performed in aortic tissues by Real Time PCR. p21-/- mice gained significantly (p < 0.01) more weight than wild type mice, triglycerides (p < 0.05) and cholesterol levels (p < 0.01) were more pronounced in the sera of p21-/- compared to wild type mice fed with high fat diet. High fat diet resulted in significantly decreased TGF-beta (p < 0.02), HO-l (p < 0.02) and increased CD36 (p < 0.03) mRNA expression in aortic tissues of p21-/- mice compared to animal fed with regular diet. IFN-gamma mRNA expression (235 +/- 11 folds) increased significantly in high fat diet fed p21-/- mice and a multifold modulation of PPAR-gamma(136 +/- 7), p22phox, NOX-1 and Rac-1 (15-35-folds) mRNA in aortic tissues from p21-/- mice compared to the wild type mice. Severity of atherosclerotic lesions was significantly higher in p21-/- compared to wild type mice. The results demonstrate that the deficiency of p21 leads to altered expression of pro-atherogenic genes, and severe atherosclerosis in mice fed with high fat diet. This opens the possibility of p21 protein as a therapeutic tool to control progression of atherosclerosis.
Collapse
Affiliation(s)
- Ashwani K Khanna
- Department of Medicine (Cardiology), University of Maryland, Baltimore, USA.
| |
Collapse
|
7
|
Bot PT, Hoefer IE, Sluijter JP, van Vliet P, Smits AM, Lebrin F, Moll F, de Vries JP, Doevendans P, Piek JJ, Pasterkamp G, Goumans MJ. Increased Expression of the Transforming Growth Factor-β Signaling Pathway, Endoglin, and Early Growth Response-1 in Stable Plaques. Stroke 2009; 40:439-47. [DOI: 10.1161/strokeaha.108.522284] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Pieter T.G. Bot
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Imo E. Hoefer
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Joost P.G. Sluijter
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Patrick van Vliet
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Anke M. Smits
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Franck Lebrin
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Frans Moll
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Jean-Paul de Vries
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Pieter Doevendans
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Jan J. Piek
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Gerard Pasterkamp
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| | - Marie-José Goumans
- From the Department of Cardiology (P.T.G.B., J.J.P.), AMC Amsterdam, The Netherlands; the Laboratory of Experimental Cardiology (P.T.G.B., I.E.H., G.P.), the Department of Cardiology (J.P.G.S., P.v.V., A.M.S., P.D., M.-J.G.), and the Department of Vascular Surgery (F.M.), UMC Utrecht, Utrecht, The Netherlands; the Department of Vascular Surgery (J.-P.d.V.), St. Antonius Hospital, Nieuwegein, The Netherlands; the Interuniversity Cardiology Institute of the Netherlands (ICIN) (J.P.G.S., P.v.V.),
| |
Collapse
|
8
|
Abstract
With cardiovascular (CV)-related disorders accounting for the highest mortality rates in the world, affecting the quantity and quality of life of patients and creating an economic burden of prolonged therapeutic intervention, there is great significance in understanding the cellular and molecular alterations that influence the progression of these pathologies. The cellular genotype is regulated by the DNA component, whilst the cellular phenotype is influenced by the protein complement. By improving the understanding of the molecular mechanisms that influence the protein profile, the pathologies that influence the intrinsic functions of the CV system may be detected earlier or managed more efficiently. This is achievable with technologies encompassed by 'proteomics.' Proteomic investigations of CV diseases, including dilated cardiomyopathy (DCM), atherosclerosis, and ischemia/reperfusion (I/R) injury, have identified candidate proteins altered with the pathologic states, complementing past biochemical and physiologic observations. Whilst proteomics is still a relatively new discipline to be applied to the basic scientific investigation of CV diseases, it is emerging as a technique to screen for potential biomarkers in both tissues/cells and biologic fluids (biofluids), as well as to identify the targets of existing therapeutics. By enabling the separation of complex mixtures over numerous dimensions, exploiting the intrinsic properties of proteins, including charge state, molecular mass, and hydrophobicity, in addition to cellular location, the discrete alterations within the cell may be resolved. Proteomics has shown alterations to myofilament proteins including troponin I and myosin light chain, correlating with the reduction in contractility in the myocardium from DCM and I/R. The diverse cell types that coalesce to induce atherosclerotic plaque formation have been investigated both collectively and individually to elucidate the influence of the modifications to single cell types on the developing plaque as a whole. Proteomics has also been used to observe changes to biofluids occurring with these pathologies, a new potential link between basic science and clinical applications. The development of CV proteomics has helped to identify a number of possible protein candidates, and offers the potential to treat and diagnose CV disease more effectively in the future.
Collapse
Affiliation(s)
- Melanie Y White
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland 21224, USA
| | | |
Collapse
|
9
|
Khanna AK, Mehra MR. Targeted in vitro and in vivo gene transfer into T lymphocytes: potential of direct inhibition of allo-immune activation. BMC Immunol 2006; 7:26. [PMID: 17096842 PMCID: PMC1657031 DOI: 10.1186/1471-2172-7-26] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Accepted: 11/10/2006] [Indexed: 11/17/2022] Open
Abstract
Background Successful inhibition of alloimmune activation in organ transplantation remains one of the key events in achieving a long-term graft survival. Since T lymphocytes are largely responsible for alloimmune activation, targeted gene transfer of gene of cyclin kinase inhibitor p21 into T cells might inhibit their aberrant proliferation. A number of strategies using either adenoviral or lentiviral vectors linked to mono or bispecific antibodies directed against T cell surface markers/cytokines did not yield the desired results. Therefore, this study was designed to test if a CD3promoter-p21 chimeric construct would in vitro and in vivo transfer p21 gene to T lymphocytes and result in inhibition of proliferation. CD3 promoter-p21 chimeric constructs were prepared with p21 in the sense and antisense orientation. For in vitro studies EL4-IL-2 thyoma cells were used and for in vivo studies CD3p21 sense and antisense plasmid DNA was injected intramuscularly in mice. Lymphocyte proliferation was quantified by 3H-thymidine uptake assay; IL-2 mRNA expression was studied by RT-PCR and using Real Time PCR assay, we monitored the CD3, p21, TNF-α and IFN-γ mRNA expression. Results Transfection of CD3p21 sense and antisense in mouse thyoma cell line (EL4-IL-2) resulted in modulation of mitogen-induced proliferation. The intramuscular injection of CD3p21 sense and antisense plasmid DNA into mice also modulated lymphocyte proliferation and mRNA expression of pro-inflammatory cytokines. Conclusion These results demonstrate a novel strategy of in vitro and in vivo transfer of p21 gene to T cells using CD3-promoter to achieve targeted inhibition of lymphocyte proliferation and immune activation.
Collapse
Affiliation(s)
- Ashwani K Khanna
- Department of Medicine, Division of Cardiology, University of Maryland, Baltimore, MD-21201 USA
| | - Mandeep R Mehra
- Department of Medicine, Division of Cardiology, University of Maryland, Baltimore, MD-21201 USA
| |
Collapse
|
10
|
Abstract
Transforming growth factor-beta (TGF-beta) superfamily members, TGF-beta and bone morphogenetic proteins (BMPs), are potent regulatory cytokines with diverse functions on vascular cells. They signal through heteromeric type I and II receptor complexes activating Smad-dependent and Smad-independent signals, which regulate proliferation, differentiation, and survival. They are potent regulators of vascular development and vessel remodeling and play key roles in atherosclerosis and restenosis, regulating endothelial, smooth muscle cell, macrophage, T cell, and probably vascular calcifying cell responses. In atherosclerosis, TGF-beta regulates lesion phenotype by controlling T-cell responses and stimulating smooth muscle cells to produce collagen. It contributes to restenosis by augmenting neointimal cell proliferation and collagen accumulation. Defective TGF-beta signaling in endothelial cells attributable to mutations in endoglin or the type I receptor ALK-1 leads to hereditary hemorrhagic telangiectasia, whereas defective BMP signaling attributable to mutations in the BMP receptor II has been associated with development of primary pulmonary hypertension. The development of mouse models with either cell type-specific or general inactivation of TGF-beta/BMP signaling has started to reveal the importance of the regulatory network of TGF-beta/BMP pathways in vivo and their significance for atherosclerosis, hereditary hemorrhagic telangiectasia, and primary pulmonary hypertension. This review highlights recent findings that have advanced our understanding of the roles of TGF-beta superfamily members in regulating vascular cell responses and provides likely avenues for future research that may lead to novel pharmacological therapies for the treatment or prevention of vascular disorders.
Collapse
Affiliation(s)
- Alex Bobik
- Cell Biology Laboratory, Baker Heart Research Institute, PO Box 6492, St Kilda Rd Central, Melbourne, Victoria 8008, Australia.
| |
Collapse
|
11
|
Zacchigna L, Vecchione C, Notte A, Cordenonsi M, Dupont S, Maretto S, Cifelli G, Ferrari A, Maffei A, Fabbro C, Braghetta P, Marino G, Selvetella G, Aretini A, Colonnese C, Bettarini U, Russo G, Soligo S, Adorno M, Bonaldo P, Volpin D, Piccolo S, Lembo G, Bressan GM. Emilin1 links TGF-beta maturation to blood pressure homeostasis. Cell 2006; 124:929-42. [PMID: 16530041 DOI: 10.1016/j.cell.2005.12.035] [Citation(s) in RCA: 222] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Revised: 11/18/2005] [Accepted: 12/14/2005] [Indexed: 11/17/2022]
Abstract
TGF-beta proteins are main regulators of blood vessel development and maintenance. Here, we report an unprecedented link between TGF-beta signaling and arterial hypertension based on the analysis of mice mutant for Emilin1, a cysteine-rich secreted glycoprotein expressed in the vascular tree. Emilin1 knockout animals display increased blood pressure, increased peripheral vascular resistance, and reduced vessel size. Mechanistically, we found that Emilin1 inhibits TGF-beta signaling by binding specifically to the proTGF-beta precursor and preventing its maturation by furin convertases in the extracellular space. In support of these findings, genetic inactivation of Emilin1 causes increased TGF-beta signaling in the vascular wall. Strikingly, high blood pressure observed in Emilin1 mutants is rescued to normal levels upon inactivation of a single TGF-beta1 allele. This study highlights the importance of modulation of TGF-beta availability in the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Luca Zacchigna
- Developmental Signaling Laboratory, Department of Histology Microbiology and Medical Biotechnologies, University of Padua, viale Colombo 3, 35121 Padua, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Johnson MC, Torres M, Alves A, Bacallao K, Fuentes A, Vega M, Boric MA. Augmented cell survival in eutopic endometrium from women with endometriosis: expression of c-myc, TGF-beta1 and bax genes. Reprod Biol Endocrinol 2005; 3:45. [PMID: 16150151 PMCID: PMC1262771 DOI: 10.1186/1477-7827-3-45] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2005] [Accepted: 09/08/2005] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Endometriosis is a common gynaecological disorder characterized by the presence of endometrial tissue outside of the uterus. The fragments in normal menstruation are composed of necrotic and living cells, which do not survive in ectopic locations because of programmed cell death. The aim of this study was to evaluate if the balance between cell proliferation and apoptosis is changed in eutopic endometrium from women with endometriosis throughout the menstrual cycle by studying bax (pro-apoptotic), c-myc (regulator of cell cycle) and TGF-beta1 (involved in cell differentiation) genes. METHODS Eutopic endometrium was obtained from: 30 women with endometriosis (32.8 +/- 5 years) and 34 fertile eumenorrheic women (36 +/- 5.3 years). We analyzed apoptosis (TUNEL: DNA fragmentation); cell proliferation (immunohistochemistry (IHC) for Ki67); c-myc, bax and TGF-beta1 mRNA abundance (RT-PCR) and TGF-beta1 protein (IHC) in endometrial explants. RESULTS Cell proliferation strongly decreased from proliferative to late secretory phases in glands, but not in stroma, in both endometria. Positive staining in glands and stroma from proliferative endometrium with endometriosis was 1.9- and 2.2-fold higher than control endometrium, respectively (p < 0.05). Abundance of c-myc mRNA was 65% higher in proliferative endometrium from endometriosis than normal tissue (p < 0.05). TGF-beta1 (mRNA and protein) augmented during mid secretory phase in normal endometrium, effect not observed in endometrium with endometriosis. In normal endometrium, the percentage of apoptotic epithelial and stromal cells increased more than 30-fold during late secretory phase. In contrast, in endometrium from endometriosis, not only this increase was not observed, besides bax mRNA decreased 63% versus normal endometrium (p < 0.05). At once, in early secretory phase, apoptotic stromal cells increased 10-fold with a concomitant augment of bax mRNA abundance (42%) in endometria from endometriosis (p < 0.05). CONCLUSION An altered expression of c-myc, TGF-beta1 and bax was observed in eutopic endometrium from endometriosis, suggesting its participation in the regulation of cell survival in this disease. The augmented cell viability in eutopic endometrium from these patients as a consequence of a reduction in cell death by apoptosis, and also an increase in cell proliferation indicates that this condition may facilitate the invasive feature of the endometrium.
Collapse
Affiliation(s)
- M Cecilia Johnson
- Institute of Maternal and Child Research, School of Medicine, University of Chile; and San Borja Arriarán Clinical Hospital, Santiago, Chile
| | - Marisa Torres
- Institute of Maternal and Child Research, School of Medicine, University of Chile; and San Borja Arriarán Clinical Hospital, Santiago, Chile
| | - Alessandra Alves
- Institute of Maternal and Child Research, School of Medicine, University of Chile; and San Borja Arriarán Clinical Hospital, Santiago, Chile
| | - Ketty Bacallao
- Institute of Maternal and Child Research, School of Medicine, University of Chile; and San Borja Arriarán Clinical Hospital, Santiago, Chile
| | - Ariel Fuentes
- Institute of Maternal and Child Research, School of Medicine, University of Chile; and San Borja Arriarán Clinical Hospital, Santiago, Chile
| | - Margarita Vega
- Institute of Maternal and Child Research, School of Medicine, University of Chile; and San Borja Arriarán Clinical Hospital, Santiago, Chile
| | - M Angélica Boric
- Institute of Maternal and Child Research, School of Medicine, University of Chile; and San Borja Arriarán Clinical Hospital, Santiago, Chile
| |
Collapse
|
13
|
Shyu KG, Chao YM, Wang BW, Kuan P. Regulation of Discoidin Domain Receptor 2 by Cyclic Mechanical Stretch in Cultured Rat Vascular Smooth Muscle Cells. Hypertension 2005; 46:614-21. [PMID: 16087782 DOI: 10.1161/01.hyp.0000175811.79863.e2] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Discoidin domain receptor 2 (DDR2) plays potential roles in the regulation of collagen turnover mediated by smooth muscle cells in atherosclerosis. How mechanical stretch affects the regulation of DDR2 in smooth muscle cells is not fully understood. We sought to investigate the cellular and molecular mechanisms of regulation of DDR2 by cyclic stretch in smooth muscle cells. Rat vascular smooth muscle cells grown on a flexible membrane base were stretched by vacuum to 20% of maximum elongation, at 60 cycles/min. Cyclic stretch significantly increased DDR2 protein and mRNA expression after stretch. Cyclic stretch also significantly increased DNA-protein binding activity of Myc-Max. Addition of SB203580, transforming growth factor-beta1 (TGF-beta1) monoclonal antibody, p38 small interfering RNA (siRNA), and c-myc siRNA 30 minutes before stretch inhibited the induction of DDR2 protein and abolished the DNA-protein binding activity induced by cyclic stretch. Cyclic stretch increased, whereas SB203580 abolished the phosphorylated p38 protein. Conditioned medium from stretched smooth muscle cells and exogenous administration of angiotensin II and TGF-beta1 recombinant proteins to the nonstretched cells increased DDR2 protein expression similar to that seen after stretch. In conclusion, cyclic mechanical stretch enhances DDR2 expression in cultured rat smooth muscle cells. The stretch-induced DDR2 is mediated by angiotensin II and TGF-beta1, at least in part, through p38 mitogen-activated protein kinase and Myc pathway.
Collapse
MESH Headings
- Angiotensin II/metabolism
- Angiotensin II/pharmacology
- Animals
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Cell Movement/physiology
- Cell Proliferation
- Cells, Cultured
- Discoidin Domain Receptors
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Proto-Oncogene Proteins c-myc/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptors, Mitogen/genetics
- Receptors, Mitogen/metabolism
- Stress, Mechanical
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/pharmacology
- Transforming Growth Factor beta1
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Kou-Gi Shyu
- Department of Education and Research, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.
| | | | | | | |
Collapse
|
14
|
Abstract
The TGF-beta superfamily is the most versatile considering the ability of its members to regulate proliferation, growth arrest, differentiation, and apoptosis of prostatic stromal and epithelial cells as well as the formation of osteoblastic metastases. TGF-beta mediated action in prostate cells follows a complex signaling pathway from binding and phosphorylation of receptor type II to the TbetaRI kinase to Smad activation, resulting in ligand-induced transcription. TGF-beta as an indirect tumor suppressor, its role of regulating tumor induction, as well as tumor suppression depending on the tissue microenvironment merits further exploration. The rationale for targeting growth factors and their receptors for therapeutic intervention is based upon the fact that these proteins represent the most proximate component of the signal transduction cascade. The alternate targeting of intracellular effectors in the signal transduction may be thwarted by cross talk between signaling pathways (such as the Smads in a dynamic interplay with the androgen receptor). TGF-beta within the context of its well-documented apoptosis regulatory actions in the prostate and the significance its key receptor TbetaRII as a potential tumor suppressor, provides a highly attractive candidate for such targeting with high clinical significance for the treatment and diagnosis of prostate cancer.
Collapse
Affiliation(s)
- Brian Zhu
- Division of Urology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | | |
Collapse
|
15
|
Zhu C, Ying D, Zhou D, Mi J, Zhang W, Chang Q, Li L. Expression of TGF-beta1 in smooth muscle cells regulates endothelial progenitor cells migration and differentiation. J Surg Res 2005; 125:151-6. [PMID: 15854667 DOI: 10.1016/j.jss.2004.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2004] [Revised: 12/09/2004] [Accepted: 12/11/2004] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Endothelial angiogenesis in the intima of the arterial wall is one of key events in the pathogenesis of arteriosclerosis. The molecular mechanisms by which transforming growth factor beta 1 (TGFbeta1) and endothelial progenitor cells may be responsible for angiogenesis of arteriosclerosis lesions are poorly understood. MATERIALS AND METHODS Primary culture smooth muscle cells were transfected with pMAMneoTGFbeta1. ELISA checked VEGF expression in smooth muscle cells. Human EPCs (CD34+ cells) were cultured in pMAMneoTGFbeta1 or pMAMneo transfected smooth muscle cells conditional medium. After 21 days, differentiated endothelial colonies were confirmed by immunofluorescence for von Willebrand factor (vWF) and vascular-endothelial (VE)-cadherin. The VEGFR-1 expression in differentiated endothelial colonies was detected by ELISA. Cells migration and adhesion toward pMAMneoTGFbeta1 and pMAMneo transfected smooth muscle cells were also measured in parallel flow chamber. RESULTS Abundant TGFbeta1 stable expressed in smooth muscle cells. TGFbeta1 transfected smooth muscle cells expressed significantly higher level VEGF than pMAMneo group. As judged by positive staining for endothelial markers vWF and VE-cadherin, the combination of TGFbeta1 transfected smooth muscle cells conditional medium produced significantly more endothelial colonies (P<0.05) than did pMAMneo group. The adhesion force between endothelial progenitor cells and smooth muscle cells in TGFbeta1 group was higher than control. CONCLUSION TGFbeta1 expressed smooth muscle cells can be helpful for increasing endothelial progenitor cells adhesion and differentiation. It may be responsible for angiogenesis of arteriosclerosis lesions and useful for blood vessel tissue engineering.
Collapse
Affiliation(s)
- Chuhong Zhu
- Department of Anatomy, Biomechanics Section under the Key Lab for Biomechanics & Tissue Engineering of Ministry of Education, Third Military Medical University, Chongqing, China.
| | | | | | | | | | | | | |
Collapse
|
16
|
Jang WG, Kim HS, Park KG, Park YB, Yoon KH, Han SW, Hur SH, Park KS, Lee IK. Analysis of proteome and transcriptome of tumor necrosis factor ? stimulated vascular smooth muscle cells with or without alpha lipoic acid. Proteomics 2004; 4:3383-93. [PMID: 15378733 DOI: 10.1002/pmic.200400972] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Vascular smooth muscle cells (VSMCs) play an important role in the development and progression of atherosclerosis. Tumor necrosis factor alpha (TNFalpha), a cytokine secreted by VSMCs and macrophages in atherosclerotic lesions, regulates a variety of cellular functions of inflammatory cells and VSMCs by promoting cell growth and motility, which are critical for the initiation and progression of vascularlesions. Alpha lipoic acid (ALA), a well known antioxidant, acts as a pyruvate dehydrogenase cofactor in mitochondrial metabolism. Recently, we reported that ALA has many beneficial effects on vascular cells in atherosclerosis. The aim of the current study was to examine VSMCs, treated for 24 hours with TNFalpha (10 ng/mL) in the presence or absence of ALA (2 mM), for differential protein and genes expression using two-dimensional gel electrophoresis (2-DE) and DNA microarray analysis, respectively. Using 2-DE, we identified proteins whose expression changed by at least 2.5-fold after TNFalpha stimulation. Proteins up-regulated by TNFalpha that were subsequently down-regulated in the presence of ALA were identified by matrix-assisted laser desorption/ionization-time of flight mass spectrometry as plasminogen activator inhibitor-2, fetal liver LKB-interacting protein, osteoblast-specific factor 2, glucosidase II, cyclin-dependent kinase 3, endoplasmin precursor and glutathione synthetase. TNFalpha down-regulated proteins that were up-regulated in the presence of ALA were keratin 19, eukaryotic translation elongation factor and Rho GDP dissociation inhibitor alpha. Gene expression analysis using DNA microarray tools confirmed the up-regulation or down-regulation of some, but not all, of the proteins observed in ALA challenged, TNFalpha-treated cells. This data should provide valuable information about the underlying mechanisms of atherosclerosis.
Collapse
Affiliation(s)
- Won Gu Jang
- Department of Genetic Engineering, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|