1
|
Park SH, Kim YP, Lee JM, Park DW, Seo JT, Gye MC. Regulation of Phosphorylation of Glycogen Synthase Kinase 3α and the Correlation with Sperm Motility in Human. World J Mens Health 2024; 42:373-383. [PMID: 37635337 PMCID: PMC10949033 DOI: 10.5534/wjmh.230004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/10/2023] [Accepted: 04/16/2023] [Indexed: 08/29/2023] Open
Abstract
PURPOSE To unravel the mechanism regulating the phosphorylation of glycogen synthase kinase 3 (GSK3) and the correlation between the inhibitory phosphorylation of GSK3α and sperm motility in human. MATERIALS AND METHODS The phosphorylation and priming phosphorylated substrate-specific kinase activity of GSK3 were examined in human spermatozoa with various motility conditions. RESULTS In human spermatozoa, GSK3α/β was localized in the head, midpiece, and principal piece of tail and p-GSK3α(Ser21) was enriched in the midpiece. The ratio of p-GSK3α(Ser21)/GSK3α was positively coupled with normal sperm motility criteria of World Health Organization. In high-motility spermatozoa, p-GSK3α(Ser21) phosphotyrosine (p-Tyr) proteins but p-GSK3α(Tyr279) markedly increased together with decreased kinase activity of GSK3 after incubation in Ca2+ containing medium. In high-motility spermatozoa, p-GSK3α(Ser21) levels were negatively coupled with kinase activity of GSK3, and which was deregulated in low-motility spermatozoa. In high-motility spermatozoa, 6-bromo-indirubin-3'-oxime, an inhibitor of kinase activity of GSK3 increased p-GSK3α(Ser21) and p-Tyr proteins. p-GSK3α(Ser21) and p-Tyr protein levels were decreased by inhibition of PKA and Akt. Calyculin A, a protein phosphatase-1/2A inhibitor, markedly increased the p-GSK3α(Ser21) and p-Tyr proteins, and significantly increased the motility of low-motility human spermatozoa. CONCLUSIONS Down regulation of kinase activity of GSK3α by inhibitory phosphorylation was positively coupled with human sperm motility, and which was regulated by Ca2+, PKA, Akt, and PP1. Small-molecule inhibitors of GSK3 and PP1 can be considered to potentiate human sperm motility.
Collapse
Affiliation(s)
- Seung Hyun Park
- Department of Life Science and Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Young-Pil Kim
- Department of Life Science and Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Jeong Min Lee
- Department of Life Science and Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Dong-Wook Park
- Graduate School of Global Pharmaceutical Industry and Clinical Pharmacy, Ajou University, Suwon, Korea
| | - Ju Tae Seo
- JTS Urology Medical Center, Seoul, Korea
| | - Myung Chan Gye
- Department of Life Science and Institute for Natural Sciences, Hanyang University, Seoul, Korea.
| |
Collapse
|
2
|
Gellhaus B, Böker KO, Schilling AF, Saul D. Therapeutic Consequences of Targeting the IGF-1/PI3K/AKT/FOXO3 Axis in Sarcopenia: A Narrative Review. Cells 2023; 12:2787. [PMID: 38132107 PMCID: PMC10741475 DOI: 10.3390/cells12242787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The high prevalence of sarcopenia in an aging population has an underestimated impact on quality of life by increasing the risk of falls and subsequent hospitalization. Unfortunately, the application of the major established key therapeutic-physical activity-is challenging in the immobile and injured sarcopenic patient. Consequently, novel therapeutic directions are needed. The transcription factor Forkhead-Box-Protein O3 (FOXO3) may be an option, as it and its targets have been observed to be more highly expressed in sarcopenic muscle. In such catabolic situations, Foxo3 induces the expression of two muscle specific ubiquitin ligases (Atrogin-1 and Murf-1) via the PI3K/AKT pathway. In this review, we particularly evaluate the potential of Foxo3-targeted gene therapy. Foxo3 knockdown has been shown to lead to increased muscle cross sectional area, through both the AKT-dependent and -independent pathways and the reduced impact on the two major downstream targets Atrogin-1 and Murf-1. Moreover, a Foxo3 reduction suppresses apoptosis, activates satellite cells, and initiates their differentiation into muscle cells. While this indicates a critical role in muscle regeneration, this mechanism might exhaust the stem cell pool, limiting its clinical applicability. As systemic Foxo3 knockdown has also been associated with risks of inflammation and cancer progression, a muscle-specific approach would be necessary. In this review, we summarize the current knowledge on Foxo3 and conceptualize a specific and targeted therapy that may circumvent the drawbacks of systemic Foxo3 knockdown. This approach presumably would limit the side effects and enable an activity-independent positive impact on skeletal muscle.
Collapse
Affiliation(s)
- Benjamin Gellhaus
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Kai O. Böker
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Arndt F. Schilling
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Dominik Saul
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72072 Tuebingen, Germany
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
3
|
Tsuchiya K. Cardiovascular complications in insulin resistance and endocrine diseases. Endocr J 2023; 70:249-257. [PMID: 36754416 DOI: 10.1507/endocrj.ej22-0457] [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] [Indexed: 02/10/2023] Open
Abstract
Cerebrovascular diseases, such as stroke and cardiovascular disease, are one of the leading causes of death in Japan. Type 2 diabetes is the most common form of diabetes and an important risk factor for these diseases. Among various pathological conditions associated with type 2 diabetes, insulin resistance has already been reported to be an important risk factor for diabetic complications. The major sites of insulin action in glucose metabolism in the body include the liver, skeletal muscle, and adipose tissue. However, insulin signaling molecules are also constitutively expressed in vascular endothelial cells, vascular smooth muscle, and monocytes/macrophages. Forkhead box class O family member proteins (FoxOs) of transcription factors play important roles in regulating glucose and lipid metabolism, oxidative stress response and redox signaling, and cell cycle progression and apoptosis. FoxOs in vascular endothelial cells strongly promote arteriosclerosis by suppressing nitric oxide production, enhancing inflammatory response, and promoting cellular senescence. In addition, primary aldosteronism and Cushing's syndrome are known to have adverse effects on the cardiovascular system, apart from hypertension, diabetes, and dyslipidemia. In the treatment of endocrine disorders, hormonal normalization by surgical treatment and receptor antagonists play an important role in preventing cardiovascular complications.
Collapse
Affiliation(s)
- Kyoichiro Tsuchiya
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
4
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
5
|
Jiang Y, Qian HY. Transcription factors: key regulatory targets of vascular smooth muscle cell in atherosclerosis. Mol Med 2023; 29:2. [PMID: 36604627 PMCID: PMC9817296 DOI: 10.1186/s10020-022-00586-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerosis (AS), leading to gradual occlusion of the arterial lumen, refers to the accumulation of lipids and inflammatory debris in the arterial wall. Despite therapeutic advances over past decades including intervention or surgery, atherosclerosis is still the most common cause of cardiovascular diseases and the main mechanism of death and disability worldwide. Vascular smooth muscle cells (VSMCs) play an imperative role in the occurrence of atherosclerosis and throughout the whole stages. In the past, there was a lack of comprehensive understanding of VSMCs, but the development of identification technology, including in vivo single-cell sequencing technology and lineage tracing with the CreERT2-loxP system, suggests that VSMCs have remarkable plasticity and reevaluates well-established concepts about the contribution of VSMCs. Transcription factors, a kind of protein molecule that specifically recognizes and binds DNA upstream promoter regions or distal enhancer DNA elements, play a key role in the transcription initiation of the coding genes and are necessary for RNA polymerase to bind gene promoters. In this review, we highlight that, except for environmental factors, VSMC genes are transcriptionally regulated through complex interactions of multiple conserved cis-regulatory elements and transcription factors. In addition, through a series of transcription-related regulatory processes, VSMCs could undergo phenotypic transformation, proliferation, migration, calcification and apoptosis. Finally, enhancing or inhibiting transcription factors can regulate the development of atherosclerotic lesions, and the downstream molecular mechanism of transcriptional regulation has also been widely studied.
Collapse
Affiliation(s)
- Yu Jiang
- grid.506261.60000 0001 0706 7839Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, 100037 China
| | - Hai-Yan Qian
- grid.506261.60000 0001 0706 7839Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, 100037 China
| |
Collapse
|
6
|
Zhao Y, Liu YS. Longevity Factor FOXO3: A Key Regulator in Aging-Related Vascular Diseases. Front Cardiovasc Med 2022; 8:778674. [PMID: 35004893 PMCID: PMC8733402 DOI: 10.3389/fcvm.2021.778674] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Forkhead box O3 (FOXO3) has been proposed as a homeostasis regulator, capable of integrating multiple upstream signaling pathways that are sensitive to environmental changes and counteracting their adverse effects due to external changes, such as oxidative stress, metabolic stress and growth factor deprivation. FOXO3 polymorphisms are associated with extreme human longevity. Intriguingly, longevity-associated single nucleotide polymorphisms (SNPs) in human FOXO3 correlate with lower-than-average morbidity from cardiovascular diseases in long-lived people. Emerging evidence indicates that FOXO3 plays a critical role in vascular aging. FOXO3 inactivation is implicated in several aging-related vascular diseases. In experimental studies, FOXO3-engineered human ESC-derived vascular cells improve vascular homeostasis and delay vascular aging. The purpose of this review is to explore how FOXO3 regulates vascular aging and its crucial role in aging-related vascular diseases.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| |
Collapse
|
7
|
Ministrini S, Puspitasari YM, Beer G, Liberale L, Montecucco F, Camici GG. Sirtuin 1 in Endothelial Dysfunction and Cardiovascular Aging. Front Physiol 2021; 12:733696. [PMID: 34690807 PMCID: PMC8527036 DOI: 10.3389/fphys.2021.733696] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/30/2021] [Indexed: 01/10/2023] Open
Abstract
Sirtuin 1 (SIRT1) is a histone deacetylase belonging to the family of Sirtuins, a class of nicotinamide adenine dinucleotide (NAD+)-dependent enzymes with multiple metabolic functions. SIRT1 localizes in the nucleus and cytoplasm, and is implicated in the regulation of cell survival in response to several stimuli, including metabolic ones. The expression of SIRT1 is associated with lifespan and is reduced with aging both in animal models and in humans, where the lack of SIRT1 is regarded as a potential mediator of age-related cardiovascular diseases. In this review, we will summarize the extensive evidence linking SIRT1 functional and quantitative defects to cellular senescence and aging, with particular regard to their role in determining endothelial dysfunction and consequent cardiovascular diseases. Ultimately, we outline the translational perspectives for this topic, in order to highlight the missing evidence and the future research steps.
Collapse
Affiliation(s)
- Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Georgia Beer
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Istituto di Ricerca e Cura a Carattere Scientifico Ospedale Policlinico San Martino Genoa–Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni G. Camici
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Wnt Signaling Pathway Proteins in Scar, Hypertrophic Scar, and Keloid: Evidence for a Continuum? Am J Dermatopathol 2021; 42:842-847. [PMID: 32310858 DOI: 10.1097/dad.0000000000001661] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Hypertrophic scars and keloids are fibroproliferative lesions characterized by excessive collagen deposition. It is unclear whether these entities represent distinct disorders or share a common pathogenesis and the molecular underpinnings of these lesions are poorly understood. Accumulating evidence suggests that the Wnt signaling pathway is a key regulator of wound healing. In this study, tissue microarray was used to evaluate the protein expression profile for Wnt3a, phosphorylated glycogen synthase kinase 3 alpha (pGSK-3α), WNT1-inducible-signaling pathway protein 1 (WISP1), and WISP2 in normal skin, scars, hypertrophic scars, and keloids. Analysis revealed significantly increased fibroblast expression of pGSK-3α in scars (27.2%), hypertrophic scars (30.4%), and keloids (57.3%) compared with normal skin (16.4%) (all differences statistically significant; P < 0.01). Analysis of WISP2 showed 94% of fibroblasts in normal skin expressing WISP2 and significantly decreased expression in scars (46.8%), hypertrophic scars (27.0%), and keloids (61.3%) (all differences statistically significant; P < 0.01). The parallel patterns of expression of pGSK-3α and WISP2 in scars and hypertrophic scars and significantly increased expression in keloids may support the notion that keloids are a truly distinct fibrosing disorder and may provide further evidence for targeting the Wnt signaling pathway in the treatment of keloids.
Collapse
|
9
|
Bai J, Liu J, Fu Z, Feng Y, Wang B, Wu W, Zhang R. Silencing lncRNA AK136714 reduces endothelial cell damage and inhibits atherosclerosis. Aging (Albany NY) 2021; 13:14159-14169. [PMID: 34015766 PMCID: PMC8202876 DOI: 10.18632/aging.203031] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 04/02/2021] [Indexed: 12/24/2022]
Abstract
Atherosclerosis correlates with ischemic cardio-cerebrovascular diseases such as coronary heart disease. Long non-coding RNAs (lncRNAs) can promote atherosclerosis. We investigated the role of the lncRNA AK136714 in atherosclerosis. Compared with the healthy group, lncRNA AK136714 expression was elevated in the plaque and plasma of the atherosclerosis patients in a GEO dataset. AK136714 silencing inhibited atherosclerosis formation in ApoE-/- mice. AK136714 silencing also protected the endothelial barrier and inhibited endothelial cell inflammation. In vitro assays showed that knockdown of AK136714 suppressed the inflammatory response and apoptosis in human umbilical vein endothelial cells (HUVECs). Moreover, AK136714 was found to bind directly to HuR to increase the mRNA stability of TNF-α, IL-1β and IL-6 mRNAs. In addition, AK136714 promoted the transcription of Bim. This study expands our understanding of the role of lncRNA AK136714 in atherosclerosis and provides potential drug targets for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jing Bai
- Department of Geriatrics, Affiliated Hospital of Hebei University of Engineering, Handan 056000, Hebei Province, China
| | - Jianxia Liu
- Department of Nursing, Affiliated Hospital of Hebei University of Engineering, Handan 056000, Hebei Province, China
| | - Zexian Fu
- Department of Scientific Research and Education, Affiliated Hospital of Hebei University of Engineering, Handan 056000, Hebei Province, China
| | - Yuanyuan Feng
- Department of Stomatology, Affiliated Hospital of Hebei University of Engineering, Handan 056000, Hebei Province, China
| | - Bing Wang
- Department of Dynamic electrocardiogram, Affiliated Hospital of Hebei University of Engineering, Handan 056000, Hebei Province, China
| | - Wenjuan Wu
- Department of Breast, The Affiliated Hospital of Hebei University of Engineering, Handan 056000, Hebei Province, China
| | - Ruiying Zhang
- Department of Geriatrics, Affiliated Hospital of Hebei University of Engineering, Handan 056000, Hebei Province, China
| |
Collapse
|
10
|
Lu X, Li H, Wang S. Hydrogen Sulfide Protects Against Uremic Accelerated Atherosclerosis via nPKCδ/Akt Signal Pathway. Front Mol Biosci 2021; 7:615816. [PMID: 33644113 PMCID: PMC7903246 DOI: 10.3389/fmolb.2020.615816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/22/2020] [Indexed: 01/09/2023] Open
Abstract
Background: Cardiovascular disease is the most common complication and leading cause of death in maintenance hemodialysis patients. Previous studies have found that disorders of cystathionine-gamma-lyase/hydrogen sulfide (CSE/H2S) system in maintenance hemodialysis patients are correlated with the risk of cardiovascular disease. Although the role of CSE/H2S system in UAAS has been preliminarily explored, the molecular mechanism of CSE/H2S is still not systematically elaborated, and the molecular mechanism of nPKCδ and its related signaling pathway in UAAS is still not thoroughly studied. Methods: Forty chronic kidney disease (CHD) patients were studied and the activation of nPKCδ in peripheral blood mononuclear cells (PBMCs) were detected. ApoE−/− mice aged 6 weeks were treated with 5/6 nephrectomy and high-fat diet to make UAAS model. They were divided into Sham group (Sham group), UAAS group (UAAS group), UAAS+L-cysteine group (UAAS+L-cys group), UAAS+sodium hydrosulfide group (UAAS+NaHS group) and UAAS+propargylglycine group (UAAS+PPG group). The UAAS+L-cys group, UAAS+NaHS group and UAAS+PPG group were respectively given L-cys, NaHS and PPG by intraperitoneal injection. The aorta was taken 6 weeks after surgery. Western blot was used to detect the activation of nPKCδ, the phosphorylation of Akt, and the expression of VCAM-1 in the aorta of mice. Results: The membrane translocation of nPKCδ in CHD patients with plaque was higher than that in CHD patients without plaque. The membrane translocation of nPKCδ and the expression of VCAM-1 in UAAS group was higher than sham group, L-cys or NaHS injection could suppress the membrane translocation of nPKCδ and the expression of VCAM-1, but PPG treatment resulted in more membrane translocation of nPKCδ and the expression of VCAM-1 (P<0.05, n=6 per group). Akt phosphorylation in UAAS group was lower than sham group, and L-cys or NaHS injection could suppress the degradation of Akt phosphorylation, but PPG treatment resulted in more decrease in the Akt phosphorylation (P<0.05, n=6 per group). Conclusion: Endogenous CSE/H2S system protected against the formation of UAAS via nPKCδ/Akt signal pathway. The imbalance of CSE/H2S system may participate in the formation of UAAS by affecting the expression of downstream molecule VCAM-1, which may be mediated by nPKCδ/Akt signaling pathway.
Collapse
Affiliation(s)
- Xiangxue Lu
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Han Li
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Shixiang Wang
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
11
|
Liu J, Wang S, Hou J, Cai H, Pan W, Dong H, Sun R, Dong H, Fang S, Yu B. Proteomics Profiling Reveals Insulin-Like Growth Factor 1, Collagen Type VI α-2 Chain, and Fermitin Family Homolog 3 as Potential Biomarkers of Plaque Erosion in ST-Segment Elevated Myocardial Infarction. Circ J 2020; 84:985-993. [PMID: 32350230 DOI: 10.1253/circj.cj-19-1206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Plaque erosion (PE) has been considered a secondary pathogenesis of ST-segment elevated myocardial infarction (STEMI) following plaque rupture (PR). Previous studies demonstrated that they had different demographic and histology characteristics and need different treatment strategy. But there are few non-invasive plasma biomarkers for distinguishing them. The present study aimed to identify non-invasive predictive biomarkers for PE and PR in patients with STEMI. METHODS AND RESULTS A total 108 patients were recruited and grouped into a PE group (n=36), a PR group (n=36), and an unstable angina pectoris (UAP) (n=36) group for analysis. A 9-plex tandem mass tag (TMT)-based proteomics was used to compare plasma protein profiles of PE, PR, and UAP. In total, 36 significant differential proteins (DPs) were identified among groups, 10 of which were screened out using bio-information analysis and validated with enzyme-linked immunosorbent assay (ELISA). The relationship of angiography and optical coherence tomography (OCT) imaging data and the 10 target DPs was analyzed statistically. Logistic regression showed elevated collagen type VI α-2 chain (COL6A2) and insulin-like growth factor 1 (IGF1), and decreased fermitin family homolog 3 (FERMT3), were positively associated with PE. Multivariate analysis indicated IGF1, FERMT3, and COL6A2 had independent predictive ability for PE. IGF1 was inversely correlated with lumen stenosis and the lipid arc of the plaque. CONCLUSIONS IGF1, COL6A2, and FERMT3 are potential predictive biomarkers of PE in STEMI patients. And IGF1 was negatively correlated with the developing of culprit plaque.
Collapse
Affiliation(s)
- Jinxin Liu
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education
| | - Shanjie Wang
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education
| | - Jingbo Hou
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education
| | - Hengxuan Cai
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education
| | - Weili Pan
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education
| | - Haimeng Dong
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University
| | - Rong Sun
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education
| | - Hui Dong
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education
| | - Shaohong Fang
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education
| | - Bo Yu
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education
| |
Collapse
|
12
|
Wang W, Wang YR, Chen J, Chen YJ, Wang ZX, Geng M, Xu DC, Wang ZY, Li JH, Xu ZD, Pan LL, Sun J. Pterostilbene Attenuates Experimental Atherosclerosis through Restoring Catalase-Mediated Redox Balance in Vascular Smooth Muscle Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:12752-12760. [PMID: 31642668 DOI: 10.1021/acs.jafc.9b05373] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Atherosclerosis, the major risk of cardiovascular events, is a chronic vascular inflammatory disease. Pterostilbene is a naturally occurring dimethylated analogue of resveratrol and has recently been demonstrated to be beneficial against cardiovascular diseases. However, the underlying mechanisms of pterostilbene on atherosclerosis remain elusive. Experimental atherosclerosis was induced by a high-fat diet (HFD) in apolipoprotein E knockout (ApoE-/-) mice. Pterostilbene was administered intragastrically for 16 weeks. We found that pterostilbene significantly attenuated thoracic and abdominal atherosclerotic plaque formation in HFD-fed ApoE-/-mice, accompanied by modulated lipid profiles and reduced production of proinflammatory cytokines (including IL-6, IFN-γ, and TNF-α). In addition, pterostilbene restored vascular redox balance in thoracic and abdominal aorta, evidenced by enhanced catalase (CAT) expression and activities, and decreased malondialdehyde and H2O2 production. Notably, pterostilbene specifically induced CAT expression and activities in the vascular smooth muscle cells (VSMCs) of thoracic and abdominal aorta. In vitro, pterostilbene markedly promoted the expression and activity of CAT and decreased ox-low-density lipoprotein (LDL)-mediated VSMC proliferation and intracellular H2O2 production, which was abolished by CAT siRNA knockdown or inhibition. Pterostilbene-induced CAT expression was associated with inhibition of Akt, PRAS40, and GSK-3β signaling activation and upregulation of PTEN. Our data clearly demonstrated that pterostilbene exerted an antiatherosclerotic effect by inducing CAT and modulating the VSMC function.
Collapse
Affiliation(s)
- Wei Wang
- School of Life Science , Hefei Normal University , Hefei 230601 , China
| | - Ya-Ru Wang
- School of Life Science , Hefei Normal University , Hefei 230601 , China
| | - Jing Chen
- School of Life Science , Hefei Normal University , Hefei 230601 , China
| | - Ya-Jun Chen
- School of Life Science , Hefei Normal University , Hefei 230601 , China
| | - Zhao-Xia Wang
- School of Life Science , Hefei Normal University , Hefei 230601 , China
| | - Ming Geng
- School of Life Science , Hefei Normal University , Hefei 230601 , China
| | - De-Cong Xu
- School of Life Science , Hefei Normal University , Hefei 230601 , China
| | - Zi-Ying Wang
- School of Life Science , Hefei Normal University , Hefei 230601 , China
| | - Jin-Hua Li
- School of Life Science , Hefei Normal University , Hefei 230601 , China
| | - Zhong-Dong Xu
- School of Life Science , Hefei Normal University , Hefei 230601 , China
| | | | | |
Collapse
|
13
|
ZHANG J, JIN J, YANG W. [Autophagy regulates the function of vascular smooth muscle cells in the formation and rupture of intracranial aneurysms]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:552-559. [PMID: 31901031 PMCID: PMC8800671 DOI: 10.3785/j.issn.1008-9292.2019.10.14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 08/24/2019] [Indexed: 06/10/2023]
Abstract
Vascular smooth muscle cells (VSMC) are the main cellular component of vessel wall. The changes of VSMC functions including phenotypic transformation and apoptosis play a critical role in the pathogenesis of intracranial aneurysm (IA). Autophagy can participate in the regulation of vascular function by regulating cell function. In the initial stage of IA, the activation of autophagy can accelerate the phenotypic transformation of VSMC and inhibit VSMC apoptosis. With the progress of IA, the relationship between autophagy and apoptosis changes from antagonism to synergy or promotion, and a large number of apoptotic VSMC lead to the rupture of IA. In this review, we describe the role of autophagy regulating the function of VSMC in the occurrence, development and rupture of IA, for further understanding the pathogenesis of IA and finding molecular targets to prevent the formation and rupture of IA.
Collapse
Affiliation(s)
| | | | - Wei YANG
- 杨巍(1976-), 男, 博士, 教授, 博士生导师, 主要从事神经生物学及药理学研究; E-mail:
;
https://orcid.org/0000-0003-3065-1843
| |
Collapse
|
14
|
Cellular Effects of Butyrate on Vascular Smooth Muscle Cells are Mediated through Disparate Actions on Dual Targets, Histone Deacetylase (HDAC) Activity and PI3K/Akt Signaling Network. Int J Mol Sci 2019; 20:ijms20122902. [PMID: 31197106 PMCID: PMC6628026 DOI: 10.3390/ijms20122902] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/08/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022] Open
Abstract
Vascular remodeling is a characteristic feature of cardiovascular diseases. Altered cellular processes of vascular smooth muscle cells (VSMCs) is a crucial component in vascular remodeling. Histone deacetylase inhibitor (HDACI), butyrate, arrests VSMC proliferation and promotes cell growth. The objective of the study is to determine the mechanism of butyrate-induced VSMC growth. Using proliferating VSMCs exposed to 5 mM butyrate, immunoblotting studies are performed to determine whether PI3K/Akt pathway that regulates different cellular effects is a target of butyrate-induced VSMC growth. Butyrate inhibits phosphorylation-dependent activation of PI3K, PDK1, and Akt, eliciting differential effects on downstream targets of Akt. Along with previously reported Ser9 phosphorylation-mediated GSK3 inactivation leading to stability, increased expression and accumulation of cyclin D1, and epigenetic histone modifications, inactivation of Akt by butyrate results in: transcriptional activation of FOXO1 and FOXO3 promoting G1 arrest through p21Cip1/Waf1 and p15INK4B upregulation; inactivation of mTOR inhibiting activation of its targets p70S6K and 4E-BP1 impeding protein synthesis; inhibition of caspase 3 cleavage and downregulation of PARP preventing apoptosis. Our findings imply butyrate abrogates Akt activation, causing differential effects on Akt targets promoting convergence of cross-talk between their complimentary actions leading to VSMC growth by arresting proliferation and inhibiting apoptosis through its effect on dual targets, HDAC activity and PI3K/Akt pathway network.
Collapse
|
15
|
Di R, Yang Z, Xu P, Xu Y. Silencing PDK1 limits hypoxia-induced pulmonary arterial hypertension in mice via the Akt/p70S6K signaling pathway. Exp Ther Med 2019; 18:699-704. [PMID: 31281449 DOI: 10.3892/etm.2019.7627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 06/15/2018] [Indexed: 12/16/2022] Open
Abstract
The present study aimed to investigate the effect of phosphoinositide-dependent protein kinase-1 (PDK1) on hypoxia-induced pulmonary arterial hypertension (PAH). A mouse model of hypoxia-induced PAH was generated using normal or PDK1-knockout mice. Histological analysis and hemodynamic evaluations were performed to identify the progression of PAH. The expression and phosphorylation of PDK1/protein kinase B (Akt) signaling pathway associated proteins were detected by western blot analysis. Increased lung vessel thickness, right ventricular (RV) systolic pressure (RVSP), RV hypertrophy index (RVHI) values [the RV weight-to-left ventricular (LV) plus septum (S) weight ratio] and PDK1 expression were observed in the hypoxia-induced PAH model compared with the normal control. The phosphorylation of AktT308, proline-rich Akt1 substrate 1 (PRAS40) and S6KT229 was also notably increased in the PAH model compared with the control. The changes of proteins were not observed in the hypoxia treated PDK1flox/+ : Tie2-Cre mice. Similarly, the RVSP and RVHI values, and PDK1 expression were reduced in the hypoxia treated PDK1flox/+: Tie2-Cre mice to a level comparable with those in the control, suggesting that PDK1 partial knockout significantly limited hypoxia-induced PAH. The results of the present study indicate that PDK1 is essential for hypoxia-induced PAH through the PDK1/Akt/S6K signaling cascades.
Collapse
Affiliation(s)
- Ruomin Di
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Zhongzhou Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, Jiangsu 210061, P.R. China
| | - Peng Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Yingjia Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
16
|
Oh S, Son M, Lee HS, Kim HS, Jeon YJ, Byun K. Protective Effect of Pyrogallol-Phloroglucinol-6,6-Bieckol from Ecklonia cava on Monocyte-Associated Vascular Dysfunction. Mar Drugs 2018; 16:E441. [PMID: 30423960 PMCID: PMC6266154 DOI: 10.3390/md16110441] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 10/27/2018] [Accepted: 11/06/2018] [Indexed: 11/16/2022] Open
Abstract
Ecklonia cava (E. cava) can alleviate vascular dysfunction in diseases associated with poor circulation. E. cava contains various polyphenols with different functions, but few studies have compared the effects of these polyphenols. Here, we comparatively investigated four major compounds present in an ethanoic extract of E. cava. These four major compounds were isolated and their effects were examined on monocyte-associated vascular inflammation and dysfunctions. Pyrogallol-phloroglucinol-6,6-bieckol (PPB) significantly inhibited monocyte migration in vitro by reducing levels of inflammatory macrophage differentiation and of its related molecular factors. In addition, PPB protected against monocyte-associated endothelial cell death by increasing the phosphorylations of PI3K-AKT and AMPK, decreasing caspase levels, and reducing monocyte-associated vascular smooth muscle cell proliferation and migration by decreasing the phosphorylations of ERK and AKT. The results of this study show that four compounds were effective for reduction of monocyte-associated vascular inflammation and dysfunctions, but PPB might be more useful for the treatment of vascular dysfunction in diseases associated with poor circulation.
Collapse
Affiliation(s)
- Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea.
| | - Myeongjoo Son
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea.
- Department of Anatomy & Cell Biology, Graduate School of Medicine, Gachon University, Incheon 21936, Korea.
| | - Hye Sun Lee
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea.
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea.
| | - Hyun-Soo Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea.
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea.
| | - Kyunghee Byun
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea.
- Department of Anatomy & Cell Biology, Graduate School of Medicine, Gachon University, Incheon 21936, Korea.
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea.
| |
Collapse
|
17
|
Vascular smooth muscle cells activate PI3K/Akt pathway to attenuate myocardial ischemia/reperfusion-induced apoptosis and autophagy by secreting bFGF. Biomed Pharmacother 2018; 107:1779-1785. [PMID: 30257397 DOI: 10.1016/j.biopha.2018.05.113] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Vascular smooth muscle cells (VSMCs) has been reported to be implicated in atherosclerotic plaque instability and rupture. Recently, it has been demonstrated that VSMCs block the progression of cardiac remodeling and thus promoting cardiac function in a rat myocardial infarction model. However, the detailed molecular mechanism of how VSMCs contributes to recovery in myocardial ischemia/reperfusion remains not fully understood. METHODS We have isolated, identified and cultured VSMCs from rats to co-culture with rat cardiomyocyte H9C2. To culture H9C2 cells under hypoxia, we utilized CoCl2-containing medium to culture for 8 h and then was replaced with normal media for additional 16 h. Cell viability was examined by MTT assay and apoptosis was determined by flow cytometry. Infarcted area of myocardial tissue was measured by TTC staining. RESULTS VSMCs was shown to promote cell viability and inhibit apoptosis of H9C2 cells under hypoxia, which exhibited upregulated anti-apoptotic protein Bcl-2 and autophagy-related protein p62, whereas pro-apoptotic protein cleaved caspase-3 and the level of LC3II/LC3I were downregulated. Then, we confirmed VSMCs played the contributory role in cell viability of H9C2 under hypoxia by secreting bFGF, which exerted its function through PI3K/Akt pathway. Finally, in vivo, the results demonstrated that VSMCs transplantation contributed to recovery of myocardial ischemia. CONCLUSION We determine that VSMCs promote recovery of infarcted cardiomyocyte through secretion of bFGF, which then activating PI3K/Akt pathway to inhibit apoptosis and autophagy. These findings provide more insights into the molecular mechanism underlying VSMCs contributing to recovery of myocardial I/R and facilitate developing therapeutical strategies for treating heart diseases.
Collapse
|
18
|
Liu Y, Ao X, Ding W, Ponnusamy M, Wu W, Hao X, Yu W, Wang Y, Li P, Wang J. Critical role of FOXO3a in carcinogenesis. Mol Cancer 2018; 17:104. [PMID: 30045773 PMCID: PMC6060507 DOI: 10.1186/s12943-018-0856-3] [Citation(s) in RCA: 339] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022] Open
Abstract
FOXO3a is a member of the FOXO subfamily of forkhead transcription factors that mediate a variety of cellular processes including apoptosis, proliferation, cell cycle progression, DNA damage and tumorigenesis. It also responds to several cellular stresses such as UV irradiation and oxidative stress. The function of FOXO3a is regulated by a complex network of processes, including post-transcriptional suppression by microRNAs (miRNAs), post-translational modifications (PTMs) and protein-protein interactions. FOXO3a is widely implicated in a variety of diseases, particularly in malignancy of breast, liver, colon, prostate, bladder, and nasopharyngeal cancers. Emerging evidences indicate that FOXO3a acts as a tumor suppressor in cancer. FOXO3a is frequently inactivated in cancer cell lines by mutation of the FOXO3a gene or cytoplasmic sequestration of FOXO3a protein. And its inactivation is associated with the initiation and progression of cancer. In experimental studies, overexpression of FOXO3a inhibits the proliferation, tumorigenic potential, and invasiveness of cancer cells, while silencing of FOXO3a results in marked attenuation in protection against tumorigenesis. The role of FOXO3a in both normal physiology as well as in cancer development have presented a great challenge to formulating an effective therapeutic strategy for cancer. In this review, we summarize the recent findings and overview of the current understanding of the influence of FOXO3a in cancer development and progression.
Collapse
Affiliation(s)
- Ying Liu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Xiang Ao
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Wei Ding
- Department of comprehensive internal medicine, Affiliated Hospital, Qingdao University, Qingdao, 266003 China
| | - Murugavel Ponnusamy
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Wei Wu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Xiaodan Hao
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Wanpeng Yu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Yifei Wang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Peifeng Li
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Jianxun Wang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| |
Collapse
|
19
|
Lee J, Lee CY, Seo HH, Bazarragchaa B, Batdelger G, Choi S, Hwang KC, Lee S, Lim S. Extract of Oxytropis pseudoglandulosa inhibits vascular smooth muscle cell proliferation and migration via suppression of ERK1/2 and Akt signaling pathways1. Clin Hemorheol Microcirc 2018; 69:277-287. [PMID: 29660921 DOI: 10.3233/ch-189126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Excessive vascular smooth muscle cell (VSMC) proliferation and migration accelerate the development of occlusive vascular disease. Therefore, finding a means to control the aberrant proliferation and migration of VSMCs has own clinical significance. In the present study, we examined the feasibility of using extract from medicinal plant Oxytropis pseudoglandulosa (OG) to control pathologic proliferation and migration of VSMCs, which never have been tested. Our data indicate that the extract of OG significantly suppressed proliferation and migration of VSMCs without cytotoxic effect, suggesting the OG extract may be an alternative agent to effectively control the aberrant VSMC proliferation and migration without any serious adverse effect. These data suggest that the extract of OG may be a potent therapeutic agent for the treatment of occlusive vascular disease and warrant further studies to identify the major acting ingredient and to validate in vivo efficacy.
Collapse
Affiliation(s)
- Jiyun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, Seoul, Korea
| | - Hyang-Hee Seo
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | | | - Gantuya Batdelger
- Institute of General and Experimental Biology, Mongolian Academy of Sciences (MAS), Ulaanbaatar, Mongolia
| | - Sangho Choi
- International Biological Material Research Center (IBMRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Korea
| |
Collapse
|
20
|
Yang X, Gong Y, He Q, Licht JD, Liaw L, Friesel RE. Loss of Spry1 attenuates vascular smooth muscle proliferation by impairing mitogen-mediated changes in cell cycle regulatory circuits. J Cell Biochem 2018; 119:3267-3279. [PMID: 29105817 PMCID: PMC5826877 DOI: 10.1002/jcb.26486] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/22/2017] [Indexed: 12/25/2022]
Abstract
Signals from growth factors or mechanical stimuli converge to promote vascular smooth muscle cell (VSMC) migration and proliferation, key events in the pathogenesis of intimal hyperplasia upon vascular injury. Spry1, a regulator of receptor tyrosine kinases (RTK), plays a role in maintaining the contractile phenotype of VSMC. The aim of the current study was to determine the role of Spry1 in VSMC proliferation in vitro and injury induced neointimal hyperplasia in vivo. VSMC proliferation and neointima formation were evaluated in cultured human aortic SMC (hAoSMC) and ligation-induced injury of mouse carotid arteries from Spry1 gene targeted mice, and their corresponding wild type littermates. Human Spry1 or non-targeting control lentiviral shRNAs were used to knock down Spry1 in hAoSMC. Time course cell cycle analysis showed a reduced fraction of S-phase cells at 12 and 24 h after growth medium stimulation in Spry1 shRNA transduced hAoSMC. Consistent with reduced S-phase entry, the induction of cyclinD1 and the levels of pRbS807/S811, pH3Ser10, and pCdc2 were also reduced, while the cell cycle inhibitor p27Kip1 was maintained in Spry1 knockdown hAoSMC. In vivo, loss of Spry1 attenuated carotid artery ligation-induced neointima formation in mice, and this effect was accompanied by a decrease in cell proliferation similar to the in vitro results. Our findings demonstrate that loss of Spry1 attenuates mitogen-induced VSMC proliferation, and thus injury-induced neointimal hyperplasia likely via insufficient activation of Akt signaling causing decreased cyclinD1 and increased p27Kip1 and a subsequent decrease in Rb and cdc2 phosphorylation.
Collapse
Affiliation(s)
- Xuehui Yang
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME
| | - Yan Gong
- Department of Biological Repositories, Wuhan University Zhongnan Hopital, Wuhan, China
| | - Qing He
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jonathan D. Licht
- Division of Hematology and Oncology
- Department of Medicine
- University of Florida Health Cancer Center
- University of Florida College of Medicine, Gainesville, FL
| | - Lucy Liaw
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME
- Department of Biological Repositories, Wuhan University Zhongnan Hopital, Wuhan, China
| | - Robert E. Friesel
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME
| |
Collapse
|
21
|
Yu H, Fellows A, Foote K, Yang Z, Figg N, Littlewood T, Bennett M. FOXO3a (Forkhead Transcription Factor O Subfamily Member 3a) Links Vascular Smooth Muscle Cell Apoptosis, Matrix Breakdown, Atherosclerosis, and Vascular Remodeling Through a Novel Pathway Involving MMP13 (Matrix Metalloproteinase 13). Arterioscler Thromb Vasc Biol 2018; 38:555-565. [PMID: 29326312 PMCID: PMC5828387 DOI: 10.1161/atvbaha.117.310502] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/02/2018] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Vascular smooth muscle cell (VSMC) apoptosis accelerates atherosclerosis and promotes breakdown of the extracellular matrix, but the mechanistic links between these 2 processes are unknown. The forkhead protein FOXO3a (forkhead transcription factor O subfamily member 3a) is activated in human atherosclerosis and induces a range of proapoptotic and other transcriptional targets. We, therefore, determined the mechanisms and consequences of FOXO3a activation in atherosclerosis and arterial remodeling after injury. APPROACH AND RESULTS Expression of a conditional FOXO3a allele (FOXO3aA3ER) potently induced VSMC apoptosis, expression and activation of MMP13 (matrix metalloproteinase 13), and downregulation of endogenous TIMPs (tissue inhibitors of MMPs). mmp13 and mmp2 were direct FOXO3a transcriptional targets in VSMCs. Activation of endogenous FOXO3a also induced MMP13, extracellular matrix degradation, and apoptosis, and MMP13-specific inhibitors and fibronectin reduced FOXO3a-mediated apoptosis. FOXO3a activation in mice with VSMC-restricted FOXO3aA3ER induced MMP13 expression and activity and medial VSMC apoptosis. FOXO3a activation in FOXO3aA3ER/ApoE-/- (apolipoprotein E deficient) mice increased atherosclerosis, increased necrotic core and reduced fibrous cap areas, and induced features of medial degeneration. After carotid artery ligation, FOXO3a activation increased VSMC apoptosis, VSMC proliferation, and neointima formation, all of which were reduced by MMP13 inhibition. CONCLUSIONS FOXO3a activation induces VSMC apoptosis and extracellular matrix breakdown, in part, because of transcriptional activation of MMP13. FOXO3a activation promotes atherosclerosis and medial degeneration and increases neointima after injury that is partly dependent on MMP13. FOXO3a-induced MMP activation represents a direct mechanistic link between VSMC apoptosis and matrix breakdown in vascular disease.
Collapse
MESH Headings
- Animals
- Apoptosis
- Atherosclerosis/enzymology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Carotid Artery Injuries/enzymology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/pathology
- Cells, Cultured
- Disease Models, Animal
- Extracellular Matrix/enzymology
- Extracellular Matrix/pathology
- Fibrosis
- Forkhead Box Protein O3/genetics
- Forkhead Box Protein O3/metabolism
- Humans
- Male
- Matrix Metalloproteinase 13/genetics
- Matrix Metalloproteinase 13/metabolism
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Knockout, ApoE
- Mice, Transgenic
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Mutation
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Necrosis
- Rats, Wistar
- Signal Transduction
- Transcriptional Activation
- Vascular Remodeling
Collapse
Affiliation(s)
- Haixiang Yu
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Adam Fellows
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Kirsty Foote
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Zhaoqing Yang
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Nichola Figg
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Trevor Littlewood
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Martin Bennett
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.).
| |
Collapse
|
22
|
4-phenylbutyrate and valproate treatment attenuates the progression of atherosclerosis and stabilizes existing plaques. Atherosclerosis 2017; 266:103-112. [PMID: 29024862 DOI: 10.1016/j.atherosclerosis.2017.09.034] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 09/13/2017] [Accepted: 09/28/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Recent evidence suggests that endoplasmic reticulum (ER) stress signaling through glycogen synthase kinase (GSK)-3α/β is involved in the activation of pro-atherosclerotic processes. In this study, we examined the effects of small molecules that interfere with ER stress-GSK3α/β signaling on the progression and regression of atherosclerosis in a mouse model. METHODS To examine atherosclerotic progression, low-density lipoprotein receptor deficient (Ldlr-/-) mice were placed on a high-fat diet (HFD) and treated with the chemical chaperone, 4-phenylbutyrate (4PBA, 3.8 g/L drinking water), or the GSK3α/β inhibitor, valproate (VPA, 625 mg VPA/kg diet), for 10 weeks. To examine potential effects on atherosclerotic regression, 4 week old Ldlr-/- mice were placed on a HFD for 16 weeks. Subsets of mice were harvested at this time or switched to a chow (low fat) diet, or a chow diet with 4PBA or VPA treatment for 4 weeks. RESULTS In the progression model, the 4PBA- and VPA-treated mice had significantly reduced lesion and necrotic core size. Treatments had no effect on metabolic parameters, including plasma and hepatic lipid levels, or plaque composition. In the regression model, mice with 4PBA or VPA treatment showed no alterations in lesion size, but the lesions had significantly smaller necrotic cores, increased vascular smooth muscle cell content, and increased collagen content. These features are consistent with more stable plaques. CONCLUSIONS The pharmacological attenuation of ER stress or inhibition of GSK3α/β impedes the development of atherosclerosis in Ldlr-/- mice and appears to promote the stabilization of existing lesions.
Collapse
|
23
|
Xin Z, Ma Z, Jiang S, Wang D, Fan C, Di S, Hu W, Li T, She J, Yang Y. FOXOs in the impaired heart: New therapeutic targets for cardiac diseases. Biochim Biophys Acta Mol Basis Dis 2017; 1863:486-498. [PMID: 27890702 DOI: 10.1016/j.bbadis.2016.11.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/24/2016] [Accepted: 11/23/2016] [Indexed: 01/17/2023]
Abstract
Cardiac diseases have a high morbidity and mortality and affect the global population. Based on recent accumulating evidence, Forkhead box O (FOXOs) play important roles in cardiac diseases. Therefore, a summary of the current literature on the molecular mechanisms and roles of FOXOs in the heart will provide valuable information. In this review, we first briefly introduce the molecular features of FOXOs. Then, we discuss the regulation and cardiac actions of the FOXO pathways. Based on this background, we expand our discussion to the roles of FOXOs in several major cardiac diseases, such as ischemic cardiac diseases, diabetic cardiomyopathy and myocardial hypertrophy. Then, we describe some methodological problems associated with the FOXO gene-modified animal models. Finally, we discuss potential future directions. The information reviewed here may be significant for the design of future studies and may increase the potential of FOXOs as therapeutic targets.
Collapse
Affiliation(s)
- Zhenlong Xin
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China.
| | - Yang Yang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
24
|
Dual effects of fructose on ChREBP and FoxO1/3α are responsible for AldoB up-regulation and vascular remodelling. Clin Sci (Lond) 2016; 131:309-325. [PMID: 28007970 DOI: 10.1042/cs20160251] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 12/04/2016] [Accepted: 12/21/2016] [Indexed: 12/24/2022]
Abstract
Increased production of methylglyoxal (MG) in vascular tissues is one of the causative factors for vascular remodelling in different subtypes of metabolic syndrome, including hypertension and insulin resistance. Fructose-induced up-regulation of aldolase B (AldoB) contributes to increased vascular MG production but the underlying mechanisms are unclear. Serum levels of MG and fructose were determined in diabetic patients with hypertension. MG level had significant positive correlations with blood pressure and fructose level respectively. C57BL/6 mice were fed with control or fructose-enriched diet for 3 months and ultrasonographic and histologic analyses were performed to evaluate arterial structural changes. Fructose-fed mice exhibited hypertension and high levels of serum MG with normal glucose level. Fructose intake increased blood vessel wall thickness and vascular smooth muscle cell (VSMC) proliferation. Western blotting and real-time PCR analysis revealed that AldoB level was significantly increased in both the aorta of fructose-fed mice and the fructose-treated VSMCs, whereas aldolase A (AldoA) expression was not changed. The knockdown of AldoB expression prevented fructose-induced MG overproduction and VSMC proliferation. Moreover, fructose significantly increased carbohydrate-responsive element-binding protein (ChREBP), phosphorylated FoxO1/3α and Akt1 levels. Fructose induced translocation of ChREBP from the cytosol to nucleus and activated AldoB gene expression, which was inhibited by the knockdown of ChREBP. Meanwhile, fructose caused FoxO1/3α shuttling from the nucleus to cytosol and inhibited its binding to AldoB promoter region. Fructose-induced AldoB up-regulation was suppressed by Akt1 inhibitor but enhanced by FoxO1/3α siRNA. Collectively, fructose activates ChREBP and inactivates FoxO1/3α pathways to up-regulate AldoB expression and MG production, leading to vascular remodelling.
Collapse
|
25
|
Chatterjee R, Chattopadhyay S, Law S. Deregulation of vital mitotic kinase-phosphatase signaling in hematopoietic stem/progenitor compartment leads to cellular catastrophe in experimental aplastic anemia. Mol Cell Biochem 2016; 422:121-134. [PMID: 27632389 DOI: 10.1007/s11010-016-2811-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/29/2016] [Indexed: 01/09/2023]
Abstract
Aplastic anemia, the paradigm of bone marrow failure, is characterized by pancytopenic peripheral blood and hypoplastic bone marrow. Among various etiologies, inappropriate use of DNA alkylating drugs like cyclophosphamide and busulfan often causes the manifestation of the dreadful disease. Cell cycle impairment in marrow hematopoietic stem/progenitor compartment together with cellular apoptosis has been recognized as culpable factors behind aplastic pathophysiologies. However, the intricate molecular mechanisms remain unrevealed till date. In the present study, we have dealt with the mechanistic intervention of the disease by peripheral blood hemogram, bone marrow histopathology, cytopathology, hematopoietic kinetic study, scanning electron microscopy, DNA damage assessment and flowcytometric analysis of cellular proliferation and apoptosis in hematopoietic stem/progenitor cell (HSPC) rich marrow compartment using busulfan and cyclophosphamidemediated mouse model. To unveil the molecular mechanisms behind aplastic pathophysiology, we further investigated the role of some crucial mitotic and apoptotic regulators like Protein kinase-B (PKB), Gsk-3β, Cyclin-D1, PP2A, Cdc25c, Plk-1, Aurora kinase-A, Chk-1 regarding the hematopoietic catastrophe. Our observations revealed that the alteration of PKB-GSK-3β axis, Plk-1, and Aurora kinase-A expressions in HSPC compartment due to DNA damage response was associated with the proliferative impairment and apoptosis during aplastic anemia. The study established the correlation between the accumulation of DNA damage and alteration of the mentioned molecules in aplastic HSPCs that lead to the hematopoietic catastrophe. We anticipate that our findings will be beneficial for developing better therapeutic strategies for the dreadful disease concerned.
Collapse
Affiliation(s)
- Ritam Chatterjee
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C.R Avenue, Kolkata, 700073, West Bengal, India
| | - Sukalpa Chattopadhyay
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C.R Avenue, Kolkata, 700073, West Bengal, India
| | - Sujata Law
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C.R Avenue, Kolkata, 700073, West Bengal, India.
| |
Collapse
|
26
|
Affiliation(s)
- Hong Lu
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington.
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington
| |
Collapse
|
27
|
Dong X, Tang S, Zhang W, Gao W, Chen Y. GPR39 activates proliferation and differentiation of porcine intramuscular preadipocytes through targeting the PI3K/AKT cell signaling pathway. J Recept Signal Transduct Res 2015; 36:130-8. [PMID: 26524639 DOI: 10.3109/10799893.2015.1056308] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The orphan G protein-coupled receptor (GPR) 39 was originally identified as the receptor of obestatin. In this study, the effects and mechanisms of GPR39 on cell proliferation and differentiation were investigated in cultured porcine intramuscular preadipocytes. METHODS Morphology of preadipocytes and accumulated lipid droplets within cells were identified by an inverted microscope. After transfected with constructed pCMV-GPR39 plasmid, cell proliferation was measured by using methyl thiazolyl tetrazolium method, mRNA expression of GPR39, CCAAT/enhancer binding protein-α (C/EBPα), peroxisome proliferator-activated receptor-γ (PPARγ), Caspase-9 and adipocyte determination and differentiation factor-1 (ADD1) was determined by RNA preparation and reverse transcription polymerase chain reaction, protein expression of phosphoinositide-3 kinase (PI3K), 3-phosphoinositide-dependent protein kinase 1, phosphorylated glycogen synthase kinase 3 (pGSK3), total Akt and phosphorylated Akt (pAkt) was analyzed by Western blot. RESULTS It found that GPR39 mRNA and protein were expressed in porcine intramuscular preadipocytes and its expression was significantly up-regulated after treatment with Zn(2+) whose function is found to be mediated by GPR39. Furthermore, over-expression of GPR39 further promoted the optical density value of cells, enhanced mRNA expression of PPARγ, C/EBPα and ADD1, and inhibited mRNA expression of Caspase-9. Protein expression of pGSK3 and pAkt was also increased by GPR39 stimulation. In addition, GPR39-induced proliferation and differentiation of porcine intramuscular preadipocytes was partially blocked by the Akt inhibitor (PDTC) and the PI3K inhibitor (LY294002). CONCLUSION It indicated that GPR39 was a transducer of Zn(2+), and enhanced proliferation and differentiation of porcine intramuscular preadipocytes through activation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Xiaoying Dong
- a College of Yingdong Agricultural Science and Engineering, Shaoguan University , Shaoguan , P.R. China
| | - Shengqiu Tang
- a College of Yingdong Agricultural Science and Engineering, Shaoguan University , Shaoguan , P.R. China
| | - Wei Zhang
- b Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Hubei Academy of Agricultural Science , Wuhan , P.R. China
| | - Weihua Gao
- c College of Animal Science, Yangtze Univeisity , Jingzhou , P.R. China , and
| | - Yanfei Chen
- d College of Yingdong Life Science, Shaoguan University , Shaoguan , P.R. China
| |
Collapse
|
28
|
Abeyrathna P, Su Y. The critical role of Akt in cardiovascular function. Vascul Pharmacol 2015; 74:38-48. [PMID: 26025205 PMCID: PMC4659756 DOI: 10.1016/j.vph.2015.05.008] [Citation(s) in RCA: 322] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/07/2015] [Accepted: 05/16/2015] [Indexed: 12/30/2022]
Abstract
Akt kinase, a member of AGC kinases, is important in many cellular functions including proliferation, migration, cell growth and metabolism. There are three known Akt isoforms which play critical and diverse roles in the cardiovascular system. Akt activity is regulated by its upstream regulatory pathways at transcriptional and post-translational levels. Beta-catenin/Tcf-4, GLI1 and Stat-3 are some of few known transcriptional regulators of AKT gene. Threonine 308 and serine 473 are the two critical phosphorylation sites of Akt1. Translocation of Akt to the cell membrane facilitates PDK1 phosphorylation of the threonine site. The serine site is phosphorylated by mTORC2. Ack1, Src, PTK6, TBK1, IKBKE and IKKε are some of the non-canonical pathways which affect the Akt activity. Protein-protein interactions of Akt to actin and Hsp90 increase the Akt activity while Akt binding to other proteins such as CTMP and TRB3 reduces the Akt activity. The action of Akt on its downstream targets determines its function in cardiovascular processes such as cell survival, growth, proliferation, angiogenesis, vasorelaxation, and cell metabolism. Akt promotes cell survival via caspase-9, YAP, Bcl-2, and Bcl-x activities. Inhibition of FoxO proteins by Akt also increases cell survival by transcriptional mechanisms. Akt stimulates cell growth and proliferation through mTORC1. Akt also increases VEGF secretion and mediates eNOS phosphorylation, vasorelaxation and angiogenesis. Akt can increase cellular metabolism through its downstream targets GSK3 and GLUT4. The alterations of Akt signaling play an important role in many cardiovascular pathological processes such as atherosclerosis, cardiac hypertrophy, and vascular remodeling. Several Akt inhibitors have been developed and tested as anti-tumor agents. They could be potential novel therapeutics for the cardiovascular diseases.
Collapse
Affiliation(s)
- Prasanna Abeyrathna
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
29
|
Uryga AK, Bennett MR. Ageing induced vascular smooth muscle cell senescence in atherosclerosis. J Physiol 2015; 594:2115-24. [PMID: 26174609 DOI: 10.1113/jp270923] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/08/2015] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis is a disease of ageing in that its incidence and prevalence increase with age. However, atherosclerosis is also associated with biological ageing, manifest by a number of typical hallmarks of ageing in the atherosclerotic plaque. Thus, accelerated biological ageing may be superimposed on the effects of chronological ageing in atherosclerosis. Tissue ageing is seen in all cells that comprise the plaque, but particularly in vascular smooth muscle cells (VSMCs). Hallmarks of ageing include evidence of cell senescence, DNA damage (including telomere attrition), mitochondrial dysfunction, a pro-inflammatory secretory phenotype, defects in proteostasis, epigenetic changes, deregulated nutrient sensing, and exhaustion of progenitor cells. In this model, initial damage to DNA (genomic, telomeric, mitochondrial and epigenetic changes) results in a number of cellular responses (cellular senescence, deregulated nutrient sensing and defects in proteostasis). Ultimately, ongoing damage and attempts at repair by continued proliferation overwhelm reparative capacity, causing loss of specialised cell functions, cell death and inflammation. This review summarises the evidence for accelerated biological ageing in atherosclerosis, the functional consequences of cell ageing on cells comprising the plaque, and the causal role that VSMC senescence plays in atherogenesis.
Collapse
Affiliation(s)
- Anna K Uryga
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Box 110, Cambridge, CB2 0QQ, UK
| | - Martin R Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Box 110, Cambridge, CB2 0QQ, UK
| |
Collapse
|
30
|
Im J, Hergert P, Nho RS. Reduced FoxO3a expression causes low autophagy in idiopathic pulmonary fibrosis fibroblasts on collagen matrices. Am J Physiol Lung Cell Mol Physiol 2015; 309:L552-61. [PMID: 26186945 DOI: 10.1152/ajplung.00079.2015] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/13/2015] [Indexed: 11/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and fatal lung disease, and fibroblasts derived from patients with IPF are resistant to type I collagen matrix-induced cell death. The alteration of the PTEN-Akt axis permits IPF fibroblasts to maintain a pathological phenotype on collagen by suppressing autophagy. However, the precise underlying mechanism by which the Akt downstream molecule suppresses autophagic activity remains elusive. FoxO3a is a direct target of Akt and is implicated with the transcriptional activation of autophagy. Therefore, we investigated whether reduced FoxO3a expression causes abnormally low autophagy in IPF fibroblasts on collagen. We found that FoxO3a mRNA and protein levels are low in IPF fibroblasts, which subsequently suppresses the autophagosomal marker LC3B expression on collagen matrix. In contrast, the majority of control fibroblasts showed an increase in FoxO3a and LC3B expression at both the mRNA and protein levels. The luciferase assay confirmed that FoxO3a binds to the promoter region of LC3B and transcriptionally activates LC3B. The overexpression of wild-type FoxO3a increased LC3B mRNA and protein expression in IPF fibroblasts, whereas the dominant negative FoxO3a decreased the LC3B level in control fibroblasts. The inhibition of autophagic activity sensitized control fibroblasts to collagen matrix-induced cell death. In contrast, enhanced viability was found when autophagic function was inhibited in IPF fibroblasts. Our study showed that aberrantly low FoxO3a expression participates in reducing autophagic activity via transcriptional suppression of LC3B in IPF fibroblasts on collagen. This suggests that low autophagic activity by the alteration of FoxO3a may contribute to IPF progression.
Collapse
Affiliation(s)
- Jintaek Im
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota; and
| | - Polla Hergert
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota; and Lung Morphology Research Core Department, University of Minnesota, Minneapolis, Minnesota
| | | |
Collapse
|
31
|
Liu K, Ying Z, Qi X, Shi Y, Tang Q. MicroRNA-1 regulates the proliferation of vascular smooth muscle cells by targeting insulin-like growth factor 1. Int J Mol Med 2015; 36:817-24. [PMID: 26166810 DOI: 10.3892/ijmm.2015.2277] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 05/29/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to investigate the role of microRNAs (miRNAs or miRs) in vascular smooth muscle cell (VSMC) proliferation and to elucidate the underlying molecular mechanisms. In a previous study, using microarray analysis, differentially expressed miRNAs were identified in primary VSMCs isolated from the medial layer of the thoracic aorta obtained from spontaneously hypertensive rats (SHRs) and Wistar Kyoto (WKY) rats. Among others, miR-1 was identified to be downregulated in VSMCs from SHRs. Thus, in the present study, we focused on miR-1, the downregulation of which was confirmed by RT-qPCR and western blot analysis in VSMCs isolated from SHRs. We identified insulin-like growth factor 1 (IGF1) as a potential target gene of miR-1, and we subsequently validated IGF1 as a target gene of miR-1 by luciferase assay. The results revealed that the exogenous overexpression of miR-1 significantly suppressed the expression of IGF1. Additionally, we demonstrated that the downregulation of IGF1 by the introduction of miR-1 attenuated the proliferation of the VSMCs, suggesting that IGF1 is a target gene of miR-1 and that the effects of miR-1 are mediated through IGF1. In conclusion, the findings of our study demonstrate that miR-1 is significantly downregulated in VSMCs and that it is an important regulator of cell proliferation. Therefore, IGF1 may be involved in the regulation of VSMC proliferation by targeting miR-1.
Collapse
Affiliation(s)
- Kun Liu
- Department of Vascular Medicine, Peking University Shougang Hospital, Beijing, P.R. China
| | - Zhang Ying
- Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Xia Qi
- Transfusion Medicine Section, Department of Clinical Laboratory of Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Ying Shi
- Beijing Youan Hospital, Beijing, P.R. China
| | - Qiang Tang
- Department of Vascular Medicine, Peking University Shougang Hospital, Beijing, P.R. China
| |
Collapse
|
32
|
Cushing L, Costinean S, Xu W, Jiang Z, Madden L, Kuang P, Huang J, Weisman A, Hata A, Croce CM, Lü J. Disruption of miR-29 Leads to Aberrant Differentiation of Smooth Muscle Cells Selectively Associated with Distal Lung Vasculature. PLoS Genet 2015; 11:e1005238. [PMID: 26020233 PMCID: PMC4447351 DOI: 10.1371/journal.pgen.1005238] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 04/26/2015] [Indexed: 12/11/2022] Open
Abstract
Differentiation of lung vascular smooth muscle cells (vSMCs) is tightly regulated during development or in response to challenges in a vessel specific manner. Aberrant vSMCs specifically associated with distal pulmonary arteries have been implicated in the pathogenesis of respiratory diseases, such as pulmonary arterial hypertension (PAH), a progressive and fatal disease, with no effective treatment. Therefore, it is highly relevant to understand the underlying mechanisms of lung vSMC differentiation. miRNAs are known to play critical roles in vSMC maturation and function of systemic vessels; however, little is known regarding the role of miRNAs in lung vSMCs. Here, we report that miR-29 family members are the most abundant miRNAs in adult mouse lungs. Moreover, high levels of miR-29 expression are selectively associated with vSMCs of distal vessels in both mouse and human lungs. Furthermore, we have shown that disruption of miR-29 in vivo leads to immature/synthetic vSMC phenotype specifically associated with distal lung vasculature, at least partially due to the derepression of KLF4, components of the PDGF pathway and ECM-related genes associated with synthetic phenotype. Moreover, we found that expression of FBXO32 in vSMCs is significantly upregulated in the distal vasculature of miR-29 null lungs. This indicates a potential important role of miR-29 in smooth muscle cell function by regulating FBXO32 and SMC protein degradation. These results are strongly supported by findings of a cell autonomous role of endogenous miR-29 in promoting SMC differentiation in vitro. Together, our findings suggested a vessel specific role of miR-29 in vSMC differentiation and function by targeting several key negative regulators. The pathogenesis of some vascular diseases, such as PAH is selectively associated with aberrant differentiation and proliferation of vSMCs of distal arteries. While significant progresses have been made in understanding the core mechanism of differentiation and proliferation of vSMCs, little is known regarding vessel specific regulations. By investigating the expression and function of miR-29 in vivo, we found a vessel selective enriched expression and function of miR-29 during mouse lung development. Interestingly, disruption of miR-29 results in defects in vSMCs differentiation of distal vessels, reminiscent of vSMC phenotype observed in the early stage of PAH in which immature/synthetic vSMCs of distal arteries failed to differentiate and were unable to tune down the expression of collagens and other extracellular-related genes. This is the first evidence that miR-29 selectively regulates vSMCs differentiation and vessel wall formation. Future implications are to study the expression and function of miR-29 in human pulmonary vascular diseases, which might lead to establishing miR-29 as a therapeutic target for disease intervention.
Collapse
Affiliation(s)
- Leah Cushing
- Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Stefan Costinean
- Department of Pathology, Ohio State Wexner Medical Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Wei Xu
- Columbia Center for Human Development, Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Columbia University, College of Physicians & Surgeons, New York, New York, United States of America
| | - Zhihua Jiang
- Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Lindsey Madden
- Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Pingping Kuang
- Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jingshu Huang
- Columbia Center for Human Development, Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Columbia University, College of Physicians & Surgeons, New York, New York, United States of America
| | - Alexandra Weisman
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Akiko Hata
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, United States of America
| | - Carlo M. Croce
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio, United States of America
| | - Jining Lü
- Columbia Center for Human Development, Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Columbia University, College of Physicians & Surgeons, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
33
|
Yu H, Littlewood T, Bennett M. Akt isoforms in vascular disease. Vascul Pharmacol 2015; 71:57-64. [PMID: 25929188 PMCID: PMC4728195 DOI: 10.1016/j.vph.2015.03.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/31/2015] [Indexed: 11/05/2022]
Abstract
The mammalian serine/threonine Akt kinases comprise three closely related isoforms: Akt1, Akt2 and Akt3. Akt activation has been implicated in both normal and disease processes, including in development and metabolism, as well as cancer and cardiovascular disease. Although Akt signalling has been identified as a promising therapeutic target in cancer, its role in cardiovascular disease is less clear. Importantly, accumulating evidence suggests that the three Akt isoforms exhibit distinct tissue expression profiles, localise to different subcellular compartments, and have unique modes of activation. Consistent with in vitro findings, genetic studies in mice show distinct effects of individual Akt isoforms on the pathophysiology of cardiovascular disease. This review summarises recent studies of individual Akt isoforms in atherosclerosis, vascular remodelling and aneurysm formation, to provide a comprehensive overview of Akt function in vascular disease.
Collapse
Affiliation(s)
- Haixiang Yu
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| | - Trevor Littlewood
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK
| | - Martin Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
34
|
Mindin regulates vascular smooth muscle cell phenotype and prevents neointima formation. Clin Sci (Lond) 2015; 129:129-45. [PMID: 25751394 DOI: 10.1042/cs20140679] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In the present study, using diverse in vitro and in vivo models, we revealed that mindin is a novel modulator of VSMC phenotype and neointima formation in an AKT-dependent manner in response to vascular injury.
Collapse
|
35
|
Rotllan N, Wanschel AC, Fernández-Hernando A, Salerno AG, Offermanns S, Sessa WC, Fernández-Hernando C. Genetic Evidence Supports a Major Role for Akt1 in VSMCs During Atherogenesis. Circ Res 2015; 116:1744-52. [PMID: 25868464 DOI: 10.1161/circresaha.116.305895] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/13/2015] [Indexed: 12/29/2022]
Abstract
RATIONALE Coronary artery disease, the direct result of atherosclerosis, is the most common cause of death in Western societies. Vascular smooth muscle cell (VSMC) apoptosis occurs during the progression of atherosclerosis and in advanced lesions and promotes plaque necrosis, a common feature of high-risk/vulnerable atherosclerotic plaques. Akt1, a serine/threonine protein kinase, regulates several key endothelial cell and VSMC functions including cell growth, migration, survival, and vascular tone. Although global deficiency of Akt1 results in impaired angiogenesis and massive atherosclerosis, the specific contribution of VSMC Akt1 remains poorly characterized. OBJECTIVE To investigate the contribution of VSMC Akt1 during atherogenesis and in established atherosclerotic plaques. METHODS AND RESULTS We generated 2 mouse models in which Akt1 expression can be suppressed specifically in VSCMs before (Apoe(-/-)Akt1(fl/fl)Sm22α(CRE)) and after (Apoe(-/-)Akt1(fl/fl)SM-MHC-CreER(T2E)) the formation of atherosclerotic plaques. This approach allows us to interrogate the role of Akt1 during the initial and late steps of atherogenesis. The absence of Akt1 in VSMCs during the progression of atherosclerosis results in larger atherosclerotic plaques characterized by bigger necrotic core areas, enhanced VSMC apoptosis, and reduced fibrous cap and collagen content. In contrast, VSMC Akt1 inhibition in established atherosclerotic plaques does not influence lesion size but markedly reduces the relative fibrous cap area in plaques and increases VSMC apoptosis. CONCLUSIONS Akt1 expression in VSMCs influences early and late stages of atherosclerosis. The absence of Akt1 in VSMCs induces features of plaque vulnerability including fibrous cap thinning and extensive necrotic core areas. These observations suggest that interventions enhancing Akt1 expression specifically in VSMCs may lessen plaque progression.
Collapse
Affiliation(s)
- Noemi Rotllan
- From the Vascular Biology and Therapeutics Program (N.R., W.C.S., C.F-.H.), Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (N.R., C.F-.H.), Department of Pharmacology (W.C.S.), Yale University School of Medicine, New Haven, CT; Leon H. Charney Division of Cardiology and Cell Biology Departments of Medicine, New York University School of Medicine, NY (A.C.W., A.F.-H., A.G.S., C.F-.H.); and Department of Pharmacology, Max-Plank-Institute for Heart and Lung Research, Bad Nauheim, Germany (S.O.)
| | - Amarylis C Wanschel
- From the Vascular Biology and Therapeutics Program (N.R., W.C.S., C.F-.H.), Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (N.R., C.F-.H.), Department of Pharmacology (W.C.S.), Yale University School of Medicine, New Haven, CT; Leon H. Charney Division of Cardiology and Cell Biology Departments of Medicine, New York University School of Medicine, NY (A.C.W., A.F.-H., A.G.S., C.F-.H.); and Department of Pharmacology, Max-Plank-Institute for Heart and Lung Research, Bad Nauheim, Germany (S.O.)
| | - Ana Fernández-Hernando
- From the Vascular Biology and Therapeutics Program (N.R., W.C.S., C.F-.H.), Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (N.R., C.F-.H.), Department of Pharmacology (W.C.S.), Yale University School of Medicine, New Haven, CT; Leon H. Charney Division of Cardiology and Cell Biology Departments of Medicine, New York University School of Medicine, NY (A.C.W., A.F.-H., A.G.S., C.F-.H.); and Department of Pharmacology, Max-Plank-Institute for Heart and Lung Research, Bad Nauheim, Germany (S.O.)
| | - Alessandro G Salerno
- From the Vascular Biology and Therapeutics Program (N.R., W.C.S., C.F-.H.), Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (N.R., C.F-.H.), Department of Pharmacology (W.C.S.), Yale University School of Medicine, New Haven, CT; Leon H. Charney Division of Cardiology and Cell Biology Departments of Medicine, New York University School of Medicine, NY (A.C.W., A.F.-H., A.G.S., C.F-.H.); and Department of Pharmacology, Max-Plank-Institute for Heart and Lung Research, Bad Nauheim, Germany (S.O.)
| | - Stefan Offermanns
- From the Vascular Biology and Therapeutics Program (N.R., W.C.S., C.F-.H.), Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (N.R., C.F-.H.), Department of Pharmacology (W.C.S.), Yale University School of Medicine, New Haven, CT; Leon H. Charney Division of Cardiology and Cell Biology Departments of Medicine, New York University School of Medicine, NY (A.C.W., A.F.-H., A.G.S., C.F-.H.); and Department of Pharmacology, Max-Plank-Institute for Heart and Lung Research, Bad Nauheim, Germany (S.O.)
| | - William C Sessa
- From the Vascular Biology and Therapeutics Program (N.R., W.C.S., C.F-.H.), Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (N.R., C.F-.H.), Department of Pharmacology (W.C.S.), Yale University School of Medicine, New Haven, CT; Leon H. Charney Division of Cardiology and Cell Biology Departments of Medicine, New York University School of Medicine, NY (A.C.W., A.F.-H., A.G.S., C.F-.H.); and Department of Pharmacology, Max-Plank-Institute for Heart and Lung Research, Bad Nauheim, Germany (S.O.)
| | - Carlos Fernández-Hernando
- From the Vascular Biology and Therapeutics Program (N.R., W.C.S., C.F-.H.), Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (N.R., C.F-.H.), Department of Pharmacology (W.C.S.), Yale University School of Medicine, New Haven, CT; Leon H. Charney Division of Cardiology and Cell Biology Departments of Medicine, New York University School of Medicine, NY (A.C.W., A.F.-H., A.G.S., C.F-.H.); and Department of Pharmacology, Max-Plank-Institute for Heart and Lung Research, Bad Nauheim, Germany (S.O.).
| |
Collapse
|
36
|
Gray K, Kumar S, Figg N, Harrison J, Baker L, Mercer J, Littlewood T, Bennett M. Effects of DNA damage in smooth muscle cells in atherosclerosis. Circ Res 2014; 116:816-26. [PMID: 25524056 DOI: 10.1161/circresaha.116.304921] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
RATIONALE DNA damage and the DNA damage response have been identified in human atherosclerosis, including in vascular smooth muscle cells (VSMCs). However, although double-stranded breaks (DSBs) are hypothesized to promote plaque progression and instability, in part, by promoting cell senescence, apoptosis, and inflammation, the direct effects of DSBs in VSMCs seen in atherogenesis are unknown. OBJECTIVE To determine the presence and effect of endogenous levels of DSBs in VSMCs on atherosclerosis. METHODS AND RESULTS Human atherosclerotic plaque VSMCs showed increased expression of multiple DNA damage response proteins in vitro and in vivo, particularly the MRE11/RAD50/NBS1 complex that senses DSB repair. Oxidative stress-induced DSBs were increased in plaque VSMCs, but DSB repair was maintained. To determine the effect of DSBs on atherosclerosis, we generated 2 novel transgenic mice lines expressing NBS1 or C-terminal deleted NBS1 only in VSMCs, and crossed them with apolipoprotein E(-/-) mice. SM22α-NBS1/apolipoprotein E(-/-) VSMCs showed enhanced DSB repair and decreased growth arrest and apoptosis, whereas SM22α-(ΔC)NBS1/apolipoprotein E(-/-) VSMCs showed reduced DSB repair and increased growth arrest and apoptosis. Accelerating or retarding DSB repair did not affect atherosclerosis extent or composition. However, VSMC DNA damage reduced relative fibrous cap areas, whereas accelerating DSB repair increased cap area and VSMC content. CONCLUSIONS Human atherosclerotic plaque VSMCs show increased DNA damage, including DSBs and DNA damage response activation. VSMC DNA damage has minimal effects on atherogenesis, but alters plaque phenotype inhibiting fibrous cap areas in advanced lesions. Inhibiting DNA damage in atherosclerosis may be a novel target to promote plaque stability.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Aorta/pathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Apolipoproteins E/deficiency
- Brachiocephalic Trunk/pathology
- Carotid Arteries/cytology
- Cell Cycle Proteins/biosynthesis
- Cell Cycle Proteins/genetics
- Cells, Cultured
- Comet Assay
- DNA Breaks, Double-Stranded
- DNA Damage
- DNA Repair Enzymes/biosynthesis
- DNA Repair Enzymes/genetics
- DNA-Binding Proteins
- Female
- Gene Expression Profiling
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microfilament Proteins/biosynthesis
- Microfilament Proteins/genetics
- Muscle Proteins/biosynthesis
- Muscle Proteins/genetics
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Nuclear Proteins/biosynthesis
- Nuclear Proteins/genetics
- Oligonucleotide Array Sequence Analysis
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Up-Regulation
Collapse
Affiliation(s)
- Kelly Gray
- From the Division of Cardiovascular Medicine (K.G., S.K., N.F., J.H., L.B., J.M., M.B.) and Department of Biochemistry (T.L.), Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Sheetal Kumar
- From the Division of Cardiovascular Medicine (K.G., S.K., N.F., J.H., L.B., J.M., M.B.) and Department of Biochemistry (T.L.), Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Nichola Figg
- From the Division of Cardiovascular Medicine (K.G., S.K., N.F., J.H., L.B., J.M., M.B.) and Department of Biochemistry (T.L.), Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - James Harrison
- From the Division of Cardiovascular Medicine (K.G., S.K., N.F., J.H., L.B., J.M., M.B.) and Department of Biochemistry (T.L.), Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Lauren Baker
- From the Division of Cardiovascular Medicine (K.G., S.K., N.F., J.H., L.B., J.M., M.B.) and Department of Biochemistry (T.L.), Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - John Mercer
- From the Division of Cardiovascular Medicine (K.G., S.K., N.F., J.H., L.B., J.M., M.B.) and Department of Biochemistry (T.L.), Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Trevor Littlewood
- From the Division of Cardiovascular Medicine (K.G., S.K., N.F., J.H., L.B., J.M., M.B.) and Department of Biochemistry (T.L.), Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Martin Bennett
- From the Division of Cardiovascular Medicine (K.G., S.K., N.F., J.H., L.B., J.M., M.B.) and Department of Biochemistry (T.L.), Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
37
|
Segedy AK, Pyle AL, Li B, Zhang Y, Babaev VR, Jat P, Fazio S, Atkinson JB, Linton MF, Young PP. Identification of small proline-rich repeat protein 3 as a novel atheroprotective factor that promotes adaptive Akt signaling in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2014; 34:2527-36. [PMID: 25278290 DOI: 10.1161/atvbaha.114.303644] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Atherosclerosis is the primary driver of cardiovascular disease, the leading cause of death worldwide. Identification of naturally occurring atheroprotective genes has become a major goal for the development of interventions that will limit atheroma progression and associated adverse events. To this end, we have identified small proline-rich repeat protein (SPRR3) as selectively upregulated in vascular smooth muscle cells (VSMCs) of atheroma-bearing arterial tissue versus healthy arterial tissue. In this study, we sought to determine the role of SPRR3 in atheroma pathophysiology. APPROACH AND RESULTS We found that atheroprone apolipoprotein E-null mice lacking SPRR3 developed significantly greater atheroma burden. To determine the cellular driver(s) of this increase, we evaluated SPRR3-dependent changes in bone marrow-derived cells, endothelial cells, and VSMCs. Bone marrow transplant of SPRR3-expressing cells into SPRR3(-/-)apolipoprotein E-deficient recipients failed to rescue atheroma burden. Similarly, endothelial cells did not exhibit a response to SPRR3 loss. However, atheromas from SPRR3-deficient mice exhibited increased TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling)-positive VSMCs compared with control. Cell death in SPRR3-deficient VSMCs was significantly increased in vitro. Conversely, SPRR3-overexpressing VSMCs exhibited reduced apoptosis compared with control. We also observed a PI3K (phosphatidylinositol 3-kinase)/Akt-dependent positive association between SPRR3 expression and levels of active Akt in VSMCs. The survival advantage seen in SPRR3-overexpressing VSMCs was abrogated after the addition of a PI3K/Akt pathway inhibitor. CONCLUSIONS These results indicate that SPRR3 protects the lesion from VSMC loss by promoting survival signaling in plaque VSMCs, thereby significantly decreasing atherosclerosis progression. As the first identified atheroma-specific VSMC prosurvival factor, SPRR3 represents a potential target for lesion-specific modulation of VSMC survival.
Collapse
Affiliation(s)
- Amanda K Segedy
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Amy L Pyle
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Bin Li
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Youmin Zhang
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Vladimir R Babaev
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Parmjit Jat
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Sergio Fazio
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - James B Atkinson
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - MacRae F Linton
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Pampee P Young
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.).
| |
Collapse
|
38
|
Abstract
Irs2-deficient mice develop type 2-like diabetes due to a reduction in β-cell mass and a failure of pancreatic islets to undergo compensatory hyperplasia in response to insulin resistance. In order to define the molecular mechanisms, we knocked down Irs2 gene expression in mouse MIN6 insulinoma cells. Insulin receptor substrate 2 (IRS2) suppression induced apoptotic cell death, which was associated with an increase in expression of the BH3-only molecule Bim. Knockdown (KD) of Bim reduced apoptotic β-cell death induced by IRS2 suppression. In Irs2-deficient mice, Bim ablation restored β-cell mass, decreased the number of TUNEL-positive cells, and restored normal glucose tolerance after glucose challenge. FoxO1 mediates Bim upregulation induced by IRS2 suppression, and FoxO1 KD partially inhibits β-cell death induced by IRS2 suppression. These results suggest that Bim plays an important role in mediating the increase in β-cell apoptosis and the reduction in β-cell mass that occurs in IRS2-deficient diabetes.
Collapse
Affiliation(s)
- Decheng Ren
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Juan Sun
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Liqun Mao
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Honggang Ye
- Department of Medicine, The University of Chicago, Chicago, IL
| | | |
Collapse
|
39
|
Tucka J, Yu H, Gray K, Figg N, Maguire J, Lam B, Bennett M, Littlewood T. Akt1 regulates vascular smooth muscle cell apoptosis through FoxO3a and Apaf1 and protects against arterial remodeling and atherosclerosis. Arterioscler Thromb Vasc Biol 2014; 34:2421-8. [PMID: 25234814 DOI: 10.1161/atvbaha.114.304284] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Vascular smooth muscle cell (VSMC) apoptosis occurs at low levels in atherosclerotic plaques and in vessel remodeling; however, the consequences and mediators of these levels are not known. Akt1 protects against VSMC apoptosis largely through inactivating target proteins such as forkhead class O transcription factor 3a (FoxO3a), but Akt1 signaling is reduced and FoxO3a activity is increased in human atherosclerosis. We therefore sought to determine whether inhibition of VSMC apoptosis via Akt1 activation regulates vessel remodeling and atherogenesis and to identify FoxO3a target proteins that mediate VSMC apoptosis. APPROACH AND RESULTS We generated mice that express an Akt1 protein that can be activated specifically in arterial VSMCs. Akt1 activation did not affect normal arteries, but inhibited VSMC apoptosis and negative remodeling after carotid ligation, indicating that VSMC apoptosis is a major determinant of vessel caliber after changes in flow. Akt1 activation inhibited VSMC apoptosis during atherogenesis and increased relative fibrous cap area in plaques. Microarray studies identified multiple FoxO3a-regulated genes involved in VSMC apoptosis, including apoptotic protease activating factor 1 as a novel target. Apoptotic protease activating factor 1 mediated the proapoptotic activity of FoxO3a, was increased in human atherosclerosis, but reduced by Akt1 activity in vivo. CONCLUSIONS Akt1 is a major regulator of VSMC survival in vivo during vessel remodeling and atherogenesis, mediated in large part through inhibition of FoxO3a and its downstream genes, including apoptotic protease activating factor 1. Our data suggest that even the low-level VSMC apoptosis seen during changes in flow determines vessel wall structure and promotes fibrous cap thinning during atherogenesis.
Collapse
Affiliation(s)
- Joanna Tucka
- From the Divisions of Cardiovascular Medicine (J.T., H.Y., K.G., N.F., M.B., T.L.), Clinical Pharmacology (J.M.), and Metabolic Research Laboratories (B.L.), University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom; and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom (T.L.)
| | - Haixiang Yu
- From the Divisions of Cardiovascular Medicine (J.T., H.Y., K.G., N.F., M.B., T.L.), Clinical Pharmacology (J.M.), and Metabolic Research Laboratories (B.L.), University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom; and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom (T.L.)
| | - Kelly Gray
- From the Divisions of Cardiovascular Medicine (J.T., H.Y., K.G., N.F., M.B., T.L.), Clinical Pharmacology (J.M.), and Metabolic Research Laboratories (B.L.), University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom; and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom (T.L.)
| | - Nichola Figg
- From the Divisions of Cardiovascular Medicine (J.T., H.Y., K.G., N.F., M.B., T.L.), Clinical Pharmacology (J.M.), and Metabolic Research Laboratories (B.L.), University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom; and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom (T.L.)
| | - Janet Maguire
- From the Divisions of Cardiovascular Medicine (J.T., H.Y., K.G., N.F., M.B., T.L.), Clinical Pharmacology (J.M.), and Metabolic Research Laboratories (B.L.), University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom; and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom (T.L.)
| | - Brian Lam
- From the Divisions of Cardiovascular Medicine (J.T., H.Y., K.G., N.F., M.B., T.L.), Clinical Pharmacology (J.M.), and Metabolic Research Laboratories (B.L.), University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom; and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom (T.L.)
| | - Martin Bennett
- From the Divisions of Cardiovascular Medicine (J.T., H.Y., K.G., N.F., M.B., T.L.), Clinical Pharmacology (J.M.), and Metabolic Research Laboratories (B.L.), University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom; and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom (T.L.)
| | - Trevor Littlewood
- From the Divisions of Cardiovascular Medicine (J.T., H.Y., K.G., N.F., M.B., T.L.), Clinical Pharmacology (J.M.), and Metabolic Research Laboratories (B.L.), University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom; and Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom (T.L.).
| |
Collapse
|
40
|
20(s)-ginsenoside Rg3 promotes apoptosis in human ovarian cancer HO-8910 cells through PI3K/Akt and XIAP pathways. Tumour Biol 2014; 35:11985-94. [PMID: 25168366 DOI: 10.1007/s13277-014-2497-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/13/2014] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer is a serious tumor which represents a great threat to women's health. Recently, researchers had found that 20(s)-ginsenoside Rg3 could inhibit growth of several cancer cell lines; however, the mechanism is not fully understood so far. In the present study, we found that 20(s)-ginsenoside Rg3 reduced cell viability and induced apoptosis in a dose- and time-dependent manner in the human ovarian cancer cells HO-8910. The induction of apoptosis was accompanied by downregulation of phosphatidylinositol 3-kinase (PI3K)/Akt family proteins and inhibitor of apoptosis protein (IAP) family proteins. 20(s)-ginsenoside Rg3 treatment resulted in activation of caspase-3 and -9, which may partly explain the anti-cancer activity of 20(s)-ginsenoside Rg3. Taken together, our study for the first time suggests that 20(s)-ginsenoside Rg3 is able to enhance apoptosis of HO-8910 cells, at least in part, through downregulation of PI3K/Akt and IAP family proteins. Moreover, the triggering of caspase-3 and -9 activation mediated apoptotic induction. Our data indicate that 20(s)-ginsenoside Rg3 is an effective apoptosis-inducing natural compound in ovarian cancer cells and may have a role in future therapies for ovarian cancer.
Collapse
|
41
|
Martínez-Hervás S, Vinué Á, Núñez L, Andrés-Blasco I, Piqueras L, Real JT, Ascaso JF, Burks DJ, Sanz MJ, González-Navarro H. Insulin resistance aggravates atherosclerosis by reducing vascular smooth muscle cell survival and increasing CX3CL1/CX3CR1 axis. Cardiovasc Res 2014; 103:324-336. [DOI: 10.1093/cvr/cvu115] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
42
|
Maxwell SA, Mousavi-Fard S. Non-Hodgkin's B-cell lymphoma: advances in molecular strategies targeting drug resistance. Exp Biol Med (Maywood) 2013; 238:971-90. [PMID: 23986223 DOI: 10.1177/1535370213498985] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Non-Hodgkin's lymphoma (NHL) is a heterogeneous class of cancers displaying a diverse range of biological phenotypes, clinical behaviours and prognoses. Standard treatments for B-cell NHL are anthracycline-based combinatorial chemotherapy regimens composed of cyclophosphamide, doxorubicin, vincristine and prednisolone. Even though complete response rates of 40-50% with chemotherapy can be attained, a substantial proportion of patients relapse, resulting in 3-year overall survival rates of about 30%. Relapsed lymphomas are refractory to subsequent treatments with the initial chemotherapy regimen and can exhibit cross-resistance to a wide variety of anticancer drugs. The emergence of acquired chemoresistance thus poses a challenge in the clinic preventing the successful treatment and cure of disseminated B-cell lymphomas. Gene-expression analyses have increased our understanding of the molecular basis of chemotherapy resistance and identified rational targets for drug interventions to prevent and treat relapsed/refractory diffuse large B-cell lymphoma. Acquisition of drug resistance in lymphoma is in part driven by the inherent genetic heterogeneity and instability of the tumour cells. Due to the genetic heterogeneity of B-cell NHL, many different pathways leading to drug resistance have been identified. Successful treatment of chemoresistant NHL will thus require the rational design of combinatorial drugs targeting multiple pathways specific to different subtypes of B-cell NHL as well as the development of personalized approaches to address patient-to-patient genetic heterogeneity. This review highlights the new insights into the molecular basis of chemorefractory B-cell NHL that are facilitating the rational design of novel strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Steve A Maxwell
- Texas A&M Health Science Center, College Station, TX 77843-1114, USA
| | | |
Collapse
|
43
|
Hoggatt AM, Kim JR, Ustiyan V, Ren X, Kalin TV, Kalinichenko VV, Herring BP. The transcription factor Foxf1 binds to serum response factor and myocardin to regulate gene transcription in visceral smooth muscle cells. J Biol Chem 2013; 288:28477-87. [PMID: 23946491 DOI: 10.1074/jbc.m113.478974] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Smooth muscle cells (SMCs) modulate their phenotype from a quiescent contractile state to a dedifferentiated, proliferative and migratory state during the pathogenesis of many diseases, including intestinal pseudoobstruction. Understanding how smooth muscle gene expression is regulated in these different phenotypic states is critical for unraveling the pathogenesis of these diseases. In the current study we examined the specific roles of Foxf1 in visceral SMC differentiation. Data show that Foxf1 is specifically required for expression of several contractile and regulatory proteins such as telokin, smooth muscle γ-actin, and Cav1.2b in visceral SMCs. Mechanistically, Foxf1 directly binds to and activates the telokin promoter. Foxf1 also directly binds to serum response factor (SRF) and myocardin-related transcription factors (MRTFs). Unlike Foxo4 and Foxq1, which bind to MRTFs and block their interaction with SRF, Foxf1 acts synergistically with these proteins to regulate telokin expression. Knock-out of Foxf1 specifically in SMCs results in neonatal lethality, with mice exhibiting GI tract abnormalities. Mice heterozygous for Foxf1 in SMC exhibited impaired colonic contractility and decreased expression of contractile proteins. These studies together with previous studies, suggest that different forkhead proteins can regulate gene expression in SMCs through modulating the activity of the SRF-myocardin axis to either promote or inhibit differentiation and proliferation thereby altering gastrointestinal contractility and development.
Collapse
Affiliation(s)
- April M Hoggatt
- From the Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202 and
| | | | | | | | | | | | | |
Collapse
|
44
|
TIAN YE, YUE XUAN, LUO DEYI, WAZIR ROMEL, WANG JIANZHONG, WU TAO, CHEN LIN, LIAO ANGHUA, WANG KUNJIE. Increased proliferation of human bladder smooth muscle cells is mediated by physiological cyclic stretch via the PI3K-SGK1-Kv1.3 pathway. Mol Med Rep 2013; 8:294-8. [DOI: 10.3892/mmr.2013.1473] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 05/07/2013] [Indexed: 11/06/2022] Open
|
45
|
Tarnawa ED, Baker MD, Aloisio GM, Carr BR, Castrillon DH. Gonadal expression of Foxo1, but not Foxo3, is conserved in diverse Mammalian species. Biol Reprod 2013; 88:103. [PMID: 23486915 DOI: 10.1095/biolreprod.112.105791] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The Foxos are key effectors of the PI3K/Akt signaling pathway and regulate diverse physiologic processes. Two of these factors, Foxo1 and Foxo3, serve specific roles in reproduction in the mouse. Foxo3 is required for suppression of primordial follicle activation in females, while Foxo1 regulates spermatogonial stem cell maintenance in males. In the mouse ovary, Foxo1 is highly expressed in somatic cells (but not in oocytes), suggesting an important functional role for Foxo1 in these cells. Given that invertebrate model species such as Caenorhabditis elegans and Drosophila melanogaster harbor a single ancestral Foxo homolog, these observations suggest that gene duplication conferred a selective advantage by permitting the Foxos to adopt distinct roles in oogenesis and spermatogenesis. Our objective was to determine if the remarkably specific expression patterns of Foxo1 and Foxo3 in mouse gonads (and, by inference, Foxo function) are conserved in diverse mammalian species. Western blotting was used to validate isoform-specific antibodies in rodents, companion animals, farm animals, nonhuman primates, and humans. Following validation of each antibody, immunohistochemistry was performed to ascertain Foxo1 and Foxo3 gonadal expression patterns. While Foxo1 expression in spermatogonia and granulosa cells was conserved in each species evaluated, Foxo3 expression in oocytes was not. Our findings suggest that Foxo3 is not uniquely required for primordial follicle maintenance in nonrodent species and that other Foxos, particularly Foxo1, may contribute to oocyte maintenance in a functionally redundant manner.
Collapse
Affiliation(s)
- Edward D Tarnawa
- Department of Obstetrics & Gynecology, Division of Reproductive Endocrinology & Infertility, University of Texas Southwestern Medical Center, Dallas, TX 75390-9072, USA
| | | | | | | | | |
Collapse
|
46
|
Zingg JM, Hasan ST, Meydani M. Molecular mechanisms of hypolipidemic effects of curcumin. Biofactors 2013; 39:101-21. [PMID: 23339042 DOI: 10.1002/biof.1072] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 10/19/2012] [Indexed: 12/14/2022]
Abstract
Recent evidence suggests potential benefits from phytochemicals and micronutrients in reducing the elevated oxidative and lipid-mediated stress associated with inflammation, obesity, and atherosclerosis. These compounds may either directly scavenge reactive oxygen or nitrogen species or they may modulate the activity of signal transduction enzymes leading to changes in the expression of antioxidant genes. Alternatively, they may reduce plasma lipid levels by modulating lipid metabolic genes in tissues and thus reduce indirectly lipid-mediated oxidative and endoplasmic reticulum stress through their hypolipidemic effect. Here we review the proposed molecular mechanisms by which curcumin, a polyphenol present in the rhizomes of turmeric (Curcuma longa) spice, influences oxidative and lipid-mediated stress in the vascular system. At the molecular level, mounting experimental evidence suggests that curcumin may act chemically as scavenger of free radicals and/or influences signal transduction (e.g., Akt, AMPK) and modulates the activity of specific transcription factors (e.g., FOXO1/3a, NRF2, SREBP1/2, CREB, CREBH, PPARγ, and LXRα) that regulate the expression of genes involved in free radicals scavenging (e.g., catalase, MnSOD, and heme oxygenase-1) and lipid homeostasis (e.g., aP2/FABP4, CD36, HMG-CoA reductase, and carnitine palmitoyltransferase-I (CPT-1)). At the cellular level, curcumin may induce a mild oxidative and lipid-metabolic stress leading to an adaptive cellular stress response by hormetic stimulation of these cellular antioxidant defense systems and lipid metabolic enzymes. The resulting lower oxidative and lipid-mediated stress may not only explain the beneficial effects of curcumin on inflammation, cardiovascular, and neurodegenerative disease, but may also contribute to the increase in maximum life-span observed in animal models.
Collapse
Affiliation(s)
- Jean-Marc Zingg
- Vascular Biology Laboratory, Jean Mayer USDA-Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA.
| | | | | |
Collapse
|
47
|
Gorenne I, Kumar S, Gray K, Figg N, Yu H, Mercer J, Bennett M. Vascular smooth muscle cell sirtuin 1 protects against DNA damage and inhibits atherosclerosis. Circulation 2012; 127:386-96. [PMID: 23224247 DOI: 10.1161/circulationaha.112.124404] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Vascular smooth muscle cells (VSMCs) in human atherosclerosis manifest extensive DNA damage and activation of the DNA damage response, a pathway that coordinates cell cycle arrest and DNA repair, or can trigger apoptosis or cell senescence. Sirtuin 1 deacetylase (SIRT1) regulates cell ageing and energy metabolism and regulates the DNA damage response through multiple targets. However, the direct role of SIRT1 in atherosclerosis and how SIRT1 in VSMCs might regulate atherosclerosis are unknown. METHODS AND RESULTS SIRT1 expression was reduced in human atherosclerotic plaques and VSMCs both derived from plaques and undergoing replicative senescence. SIRT1 inhibition reduced DNA repair and induced apoptosis, in part, through reduced activation of the repair protein Nijmegen Breakage Syndrome-1 but not p53. Fat feeding reduced SIRT1 and induced DNA damage in VSMCs. VSMCs from mice expressing inactive truncated SIRT1 (Δex4) showed increased oxidized low-density lipoprotein-induced DNA damage and senescence. ApoE(-/-) mice expressing SIRT1(Δex4) only in smooth muscle cells demonstrated increased DNA damage response activation and apoptosis, increased atherosclerosis, reduced relative fibrous cap thickness, and medial degeneration. CONCLUSIONS SIRT1 is reduced in human atherosclerosis and is a critical regulator of the DNA damage response and survival in VSMCs. VSMC SIRT1 protects against DNA damage, medial degeneration, and atherosclerosis.
Collapse
|
48
|
Dobashi Y, Kimura M, Matsubara H, Endo S, Inazawa J, Ooi A. Molecular alterations in AKT and its protein activation in human lung carcinomas. Hum Pathol 2012; 43:2229-40. [DOI: 10.1016/j.humpath.2012.03.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 03/09/2012] [Accepted: 03/15/2012] [Indexed: 10/28/2022]
|
49
|
Dionyssiou MG, Nowacki NB, Hashemi S, Zhao J, Kerr A, Tsushima RG, McDermott JC. Cross-talk between glycogen synthase kinase 3β (GSK3β) and p38MAPK regulates myocyte enhancer factor 2 (MEF2) activity in skeletal and cardiac muscle. J Mol Cell Cardiol 2012; 54:35-44. [PMID: 23137781 DOI: 10.1016/j.yjmcc.2012.10.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 10/26/2012] [Accepted: 10/28/2012] [Indexed: 12/21/2022]
Abstract
Characterizing the signaling network that controls MEF2 transcription factors is crucial for understanding skeletal and cardiac muscle gene expression. Glycogen synthase kinase 3β (GSK3β) regulates MEF2 activity indirectly through reciprocal regulation of p38MAPK. Cross-talk between GSK3β and p38MAPK regulates MEF2 activity in skeletal and cardiac muscle. Understanding cross-talk in the signaling network converging at MEF2 control has therapeutic implications in cardiac and skeletal muscle pathology. Glycogen synthase kinase 3β (GSK3β) is a known regulator of striated muscle gene expression suppressing both myogenesis and cardiomyocyte hypertrophy. Since myocyte enhancer factor 2 (MEF2) proteins are key transcriptional regulators in both systems, we assessed whether MEF2 is a target for GSK3β. Pharmacological inhibition of GSK3β resulted in enhanced MEF2A/D expression and transcriptional activity in skeletal myoblasts and cardiac myocytes. Even though in silico analysis revealed GSK3β consensus (S/T)XXX(S/T) sites on MEF2A, a subsequent in vitro kinase assay revealed that MEF2A is only a weak substrate. However, we did observe a posttranslational modification in MEF2A in skeletal myoblasts treated with a GSK3β inhibitor which coincided with increased p38MAPK phosphorylation, a potent MEF2A activator, indicating that GSK3β inhibition may de-repress p38MAPK. Heart specific excision of GSK3β in mice also resulted in up-regulation of p38MAPK activity. Interestingly, upon pharmacological p38MAPK inhibition (SB203580), GSK3β inhibition loses its effect on MEF2 transcriptional activity suggesting potent cross-talk between the two pathways. Thus we have documented that cross-talk between p38MAPK and GSK3β signaling converges on MEF2 activity having potential consequences for therapeutic modulation of cardiac and skeletal muscle gene expression.
Collapse
Affiliation(s)
- M G Dionyssiou
- Department of Biology, York University, 4700 Keele St., Toronto, Ontario, Canada M3J 1P3
| | | | | | | | | | | | | |
Collapse
|
50
|
Lee GL, Chang YW, Wu JY, Wu ML, Wu KK, Yet SF, Kuo CC. TLR 2 induces vascular smooth muscle cell migration through cAMP response element-binding protein-mediated interleukin-6 production. Arterioscler Thromb Vasc Biol 2012; 32:2751-60. [PMID: 22995520 DOI: 10.1161/atvbaha.112.300302] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Migration of vascular smooth muscle cells (VSMCs) from the media into intima contributes to the development of atherosclerosis. Gene deletion experiments implicate a role for toll-like receptor 2 (TLR2) in atherogenesis. However, the underlying mechanisms remain unclear. We postulate that TLR2 promotes VSMC migration by enhancing interleukin (IL)-6 production. METHODS AND RESULTS Migration assays revealed that TLR2 agonists promoted VSMC migration but not cell proliferation or viability. TLR2 deficiency or inhibition of TLR2 signaling with anti-TLR2 antibody suppressed TLR2 agonist-induced VSMC migration and IL-6 production, which was mediated via p38 mitogen-associated protein kinase and extracellular signal-regulated kinase 1/2 signaling pathways. Neutralizing anti-IL-6 antibodies impaired TLR2-mediated VSMC migration and formation of filamentous actin fiber and lamellipodia. Blockade of p38 mitogen-associated protein kinase or extracellular signal-regulated kinase 1/2 activation inhibited TLR2 agonist pam3CSK4-induced phosphorylation of cAMP response element-binding protein, which regulates IL-6 promoter activity through the cAMP response element site. Moreover, cAMP response element-binding protein small interfering RNA inhibited pam3CSK4-induced IL-6 production and VSMC migration. Additionally, Rac1 small interfering RNA inhibited pam3CSK4-induced VSMC migration but not IL-6 production. CONCLUSIONS Our results suggest that on ligand binding, TLR2 activates p38 mitogen-associated protein kinase and extracellular signal-regulated kinase 1/2 signaling in VSMCs. These signaling pathways act in concert to activate cAMP response element-binding protein and subsequent IL-6 production, which in turn promotes VSMC migration via Rac1-mediated actin cytoskeletal reorganization.
Collapse
Affiliation(s)
- Guan-Lin Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
| | | | | | | | | | | | | |
Collapse
|