1
|
Ghafouri-Fard S, Khoshbakht T, Hussen BM, Taheri M, Samsami M. Emerging role of non-coding RNAs in the regulation of Sonic Hedgehog signaling pathway. Cancer Cell Int 2022; 22:282. [PMID: 36100906 PMCID: PMC9469619 DOI: 10.1186/s12935-022-02702-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/04/2022] [Indexed: 12/04/2022] Open
Abstract
Sonic Hedgehog (Shh) signaling cascade is one of the complex signaling pathways that control the accurately organized developmental processes in multicellular organisms. This pathway has fundamental roles in the tumor formation and induction of resistance to conventional therapies. Numerous non-coding RNAs (ncRNAs) have been found to interact with Shh pathway to induce several pathogenic processes, including malignant and non-malignant disorders. Many of the Shh-interacting ncRNAs are oncogenes whose expressions have been increased in diverse malignancies. A number of Shh-targeting miRNAs such as miR-26a, miR-1471, miR-129-5p, miR-361-3p, miR-26b-5p and miR-361-3p have been found to be down-regulated in tumor tissues. In addition to malignant conditions, Shh-interacting ncRNAs can affect tissue regeneration and development of neurodegenerative disorders. XIST, LOC101930370, lncRNA-Hh, circBCBM1, SNHG6, LINC‐PINT, TUG1 and LINC01426 are among long non-coding RNAs/circular RNAs that interact with Shh pathway. Moreover, miR-424, miR-26a, miR-1471, miR-125a, miR-210, miR-130a-5p, miR-199b, miR-155, let-7, miR-30c, miR-326, miR-26b-5p, miR-9, miR-132, miR-146a and miR-425-5p are among Shh-interacting miRNAs. The current review summarizes the interactions between ncRNAs and Shh in these contexts.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayyebeh Khoshbakht
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq.,Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region,, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany. .,Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Majid Samsami
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Cao F, Wang C, Long D, Deng Y, Mao K, Zhong H. Network-Based Integrated Analysis of Transcriptomic Studies in Dissecting Gene Signatures for LPS-Induced Acute Lung Injury. Inflammation 2021; 44:2486-2498. [PMID: 34462829 PMCID: PMC8405180 DOI: 10.1007/s10753-021-01518-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/07/2021] [Indexed: 10/26/2022]
Abstract
Acute lung injury (ALI) is a type of serious clinical syndrome leading to morbidity and mortality. However, the precise pathogenesis of ALI remains elusive. Here, we implemented an integrative meta-analysis of six GEO microarray studies with 76 samples in the ALI mouse model. A total of 958 differentially expressed genes (DEGs) were identified in LPS relative to normal samples. Then, a network-based meta-analysis was used to mine core DEGs and to unfold the interactions among these genes. We found that Ebi3 was the top upregulated genes in the LPS-induced ALI. GO, KEGG, and GSEA analyses were performed for functional annotation. qRT-PCR revealed augmented expression of six candidate genes (Stat1, Syk, Jak3, Rac2, Ripk1, and Traf6) in the established ALI mouse model with LPS exposure. Taken together, our study investigated comprehensively hub DEGs and their networks for LPS-stimulated ALI, which might afford an additional approach to determine biomarkers and therapeutic targets and explore the molecular pathophysiology toward ALI.
Collapse
Affiliation(s)
- Fang Cao
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, Huichuan District, 149 Dalian Road, Zunyi, Guizhou, 563003 China
| | - Chunyan Wang
- Department of Gastroenterology, Sichuan Provincial Peoples Hospital, University of Electronic Science and Technology, Chengdu, 610000 Sichuan China
| | - Danling Long
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 Hubei China
| | - Yujuan Deng
- School of Computer Science and Engineering, Shijiazhuang University, Shijiazhuang, Hebei China
| | - Kaimin Mao
- Department of Critical Care Medicine, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, 200127 China
| | - Hua Zhong
- College of Life Sciences, Wuhan University, Wuhan, 430072 Hubei China
| |
Collapse
|
3
|
Pereira-Figueiredo D, Nascimento AA, Cunha-Rodrigues MC, Brito R, Calaza KC. Caffeine and Its Neuroprotective Role in Ischemic Events: A Mechanism Dependent on Adenosine Receptors. Cell Mol Neurobiol 2021; 42:1693-1725. [PMID: 33730305 DOI: 10.1007/s10571-021-01077-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Ischemia is characterized by a transient, insufficient, or permanent interruption of blood flow to a tissue, which leads to an inadequate glucose and oxygen supply. The nervous tissue is highly active, and it closely depends on glucose and oxygen to satisfy its metabolic demand. Therefore, ischemic conditions promote cell death and lead to a secondary wave of cell damage that progressively spreads to the neighborhood areas, called penumbra. Brain ischemia is one of the main causes of deaths and summed with retinal ischemia comprises one of the principal reasons of disability. Although several studies have been performed to investigate the mechanisms of damage to find protective/preventive interventions, an effective treatment does not exist yet. Adenosine is a well-described neuromodulator in the central nervous system (CNS), and acts through four subtypes of G-protein-coupled receptors. Adenosine receptors, especially A1 and A2A receptors, are the main targets of caffeine in daily consumption doses. Accordingly, caffeine has been greatly studied in the context of CNS pathologies. In fact, adenosine system, as well as caffeine, is involved in neuroprotection effects in different pathological situations. Therefore, the present review focuses on the role of adenosine/caffeine in CNS, brain and retina, ischemic events.
Collapse
Affiliation(s)
- D Pereira-Figueiredo
- Neurobiology of the Retina Laboratory, Biomedical Sciences Program, Biomedical Institute, Fluminense Federal University, Niterói, RJ, Brazil
| | - A A Nascimento
- Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - M C Cunha-Rodrigues
- Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - R Brito
- Laboratory of Neuronal Physiology and Pathology, Cellular and Molecular Biology Department, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - K C Calaza
- Neurobiology of the Retina Laboratory, Biomedical Sciences Program, Biomedical Institute, Fluminense Federal University, Niterói, RJ, Brazil. .,Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil. .,Neurobiology Department, Biology Institute of Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
4
|
Perez JET, Ortiz-Urbina J, Heredia CP, Pham TT, Madala S, Hartley CJ, Entman ML, Taffet GE, Reddy AK. Aortic acceleration as a noninvasive index of left ventricular contractility in the mouse. Sci Rep 2021; 11:536. [PMID: 33436716 PMCID: PMC7804023 DOI: 10.1038/s41598-020-79866-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/11/2020] [Indexed: 11/09/2022] Open
Abstract
The maximum value of the first derivative of the invasively measured left ventricular (LV) pressure (+ dP/dtmax or P') is often used to quantify LV contractility, which in mice is limited to a single terminal study. Thus, determination of P' in mouse longitudinal/serial studies requires a group of mice at each desired time point resulting in "pseudo" serial measurements. Alternatively, a noninvasive surrogate for P' will allow for repeated measurements on the same group of mice, thereby minimizing physiological variability and requiring fewer animals. In this study we evaluated aortic acceleration and other parameters of aortic flow velocity as noninvasive indices of LV contractility in mice. We simultaneously measured LV pressure invasively with an intravascular pressure catheter and aortic flow velocity noninvasively with a pulsed Doppler probe in mice, at baseline and after the administration of the positive inotrope, dobutamine. Regression analysis of P' versus peak aortic velocity (vp), peak velocity squared/rise time (vp2/T), peak (+ dvp/dt or v'p) and mean (+ dvm/dt or v'm) aortic acceleration showed a high degree of association (P' versus: vp, r2 = 0.77; vp2/T, r2 = 0.86; v'p, r2 = 0.80; and v'm, r2 = 0.89). The results suggest that mean or peak aortic acceleration or the other parameters may be used as a noninvasive index of LV contractility.
Collapse
Affiliation(s)
- Jorge Enrique Tovar Perez
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, MS:BCM285, Houston, TX, 77030, USA
- Texas A&M University, Houston, TX, USA
| | - Jesus Ortiz-Urbina
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, MS:BCM285, Houston, TX, 77030, USA
| | - Celia Pena Heredia
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, MS:BCM285, Houston, TX, 77030, USA
- Houston Methodist Hospital, Houston, TX, USA
| | - Thuy T Pham
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, MS:BCM285, Houston, TX, 77030, USA
| | - Sridhar Madala
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, MS:BCM285, Houston, TX, 77030, USA
- Indus Instruments, Webster, TX, USA
| | - Craig J Hartley
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, MS:BCM285, Houston, TX, 77030, USA
| | - Mark L Entman
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, MS:BCM285, Houston, TX, 77030, USA
| | - George E Taffet
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, MS:BCM285, Houston, TX, 77030, USA
- Houston Methodist Hospital, Houston, TX, USA
| | - Anilkumar K Reddy
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, MS:BCM285, Houston, TX, 77030, USA.
- Indus Instruments, Webster, TX, USA.
| |
Collapse
|
5
|
Lin Z, Yang F, Lu D, Sun W, Zhu G, Lan B. Knockdown of NCOA2 Inhibits the Growth and Progression of Gastric Cancer by Affecting the Wnt Signaling Pathway-Related Protein Expression. Technol Cancer Res Treat 2021; 19:1533033820928072. [PMID: 32489143 PMCID: PMC7273340 DOI: 10.1177/1533033820928072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Objective: The aim of the study is to determine the role of nuclear receptor coactivator
2 in cell proliferation and invasion ability of gastric cancer cells and to
explore its possible mechanisms. Methods: Immunohistochemical staining was used to determine NCOA2
gene expression in gastric cancer. Western blotting was used to detect Wnt
signal pathways–related protein expression. Colony formation assays, Cell
Counting Kit-8 assays, and transwell assays were used to determine cell
proliferation, metastasis, and invasion ability of gastric cancer cells. A
flow cytometric apoptosis tests determine gastric cancer cell apoptosis
ability after inhibition of the expression of nuclear receptor coactivator
2. Subcutaneous mouse models were used to determine the gastric cancer
growth and peritoneal metastasis differences after inhibition the expression
of nuclear receptor coactivator 2. Results: The expression of nuclear receptor coactivator 2 in gastric cancer cells is
high (P < .01), including lymph node metastasis, TNM
staging, and gender differences in nuclear receptor coactivator 2 expression
were statistically significant (P < .01). Short
interfering nuclear receptor coactivator 2 could inhibit the proliferation
and invasion ability of gastric cancer cells. Short interfering nuclear
receptor coactivator 2 promotes the apoptosis of gastric cancer cells.
Animal experiments showed that short interfering nuclear receptor
coactivator 2 could inhibit the growth and invasion of gastric
cancer-transplantable tumors. Knockdown of the expression of nuclear
receptor coactivator 2 inhibited the Wnt/β-catenin signaling pathway in the
gastric cancer cells. Conclusions: Knockdown of the expression of nuclear receptor coactivator 2 can inhibit the
proliferation and invasion of human gastric cancer in vitro
and in vivo. The underlying mechanism of NOCA2 affects the
Wnt signaling pathway.
Collapse
Affiliation(s)
- Zhenlv Lin
- Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Fan Yang
- Department of Pediatric Surgery, First Hospital of Quanzhou, Fujian Medical University, Quanzhou, China
| | - Dong Lu
- Department of Gastrointestinal Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Wenjie Sun
- Department of Gastroenterological Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guangwei Zhu
- Department of Gastroenterological Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Bin Lan
- Department of Gastroenterological Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
6
|
Chasapi A, Balampanis K, Tanoglidi A, Kourea E, Lambrou GI, Lambadiari V, Kalfarentzos F, Hatziagelaki E, Melachrinou M, Sotiropoulou-Bonikou G. SRC-3/AIB-1 may Enhance Hepatic NFATC1 Transcription and Mediate Inflammation in a Tissue-Specific Manner in Morbid Obesity. Endocr Metab Immune Disord Drug Targets 2019; 20:242-255. [PMID: 31322077 DOI: 10.2174/1871530319666190715160630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Obesity is a global epidemic which is associated with several cardiometabolic comorbidities and is characterized by chronic, low grade systemic inflammation. Numerous biomarkers have been implicated in the pathophysiology of the disease, including transcription factors and coregulators. Steroid Receptor Coactivator (SRC)-family represent the master regulators of metabolic pathways and their dysregulation is strongly associated with numerous metabolic disorders. METHODS 50 morbidly obese patients participated in the present study. Biopsies were collected from visceral adipose tissue, subcutaneous adipose tissue, skeletal muscle, extra-myocellular adipose tissue and liver. We evaluated the differential protein expression of NFATc1, SRC-2/TIF-2, SRC-3/AIB-1 and inflammatory biomarkers CD68 and CD3 by immunohistochemistry. The current study was designed to determine any correlations between the transcription factor NFATc1 and the SRC coregulators, as well as any associations with the inflammatory biomarkers. RESULTS We identified SRC-3 as a hepatic NFATc1 coactivator and we demonstrated its possible role in energy homeostasis and lipid metabolism. Moreover, we revealed a complex and extensive intraand inter-tissue network among the three main investigated proteins and the inflammatory biomarkers, suggesting their potential participation in the obesity-induced inflammatory cascade. CONCLUSION Steroid receptor coactivators are critical regulators of human metabolism with pleiotropic and tissue-specific actions. We believe that our study will contribute to the better understanding of the complex multi-tissue interactions that are disrupted in obesity and can therefore lead to numerous cardiometabolic diseases. Further on, our present findings suggest that SRC-3/AIB-1 could constitute possible future drug targets.
Collapse
Affiliation(s)
- Athina Chasapi
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece
| | - Konstantinos Balampanis
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.,Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.,Department of Anatomy and Histology-Embryology, Medical School, University of Patras, 26500 Patras, Greece
| | - Anna Tanoglidi
- Department of Clinical Pathology, Akademiska University, Uppsala, Sweden
| | - Eleni Kourea
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece
| | - George I Lambrou
- First Department of Pediatrics, Choremeio Research Laboratory, National and Kapodistrian University of Athens, Medical School, Thivon & Levadeias 8, 11527, Goudi, Athens, Greece
| | - Vaia Lambadiari
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece
| | - Fotios Kalfarentzos
- First Department of Propaedeutic Medicine, National and Kapodistrian University of Athens Medical School, Laiko General Hospital, 17, Ag. Thoma St, 11527 Athens, Greece
| | - Erifili Hatziagelaki
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece
| | - Maria Melachrinou
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece
| | | |
Collapse
|
7
|
Quan XJ, Liang CL, Sun MZ, Zhang L, Li XL. Overexpression of steroid receptor coactivators alleviates hyperglycemia-induced endothelial cell injury in rats through activating the PI3K/Akt pathway. Acta Pharmacol Sin 2019; 40:648-657. [PMID: 30089865 PMCID: PMC6786429 DOI: 10.1038/s41401-018-0109-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 07/04/2018] [Indexed: 11/09/2022] Open
Abstract
Hyperglycemia is a major factor in vascular endothelial injury that finally leads to a cardiovascular event. Steroid receptor coactivators (SRCs) are a group of non-DNA binding proteins that induce structural changes in steroid receptors (nuclear receptors) critical for transcriptional activation. SRCs, namely, SRC-1, SRC-2, and SRC-3, are implicated in the regulation of vascular homeostasis. In this study we investigate the role of SRCs in hyperglycemia-induced endothelial injury. Aortic endothelial cells were prepared from normal and diabetic rats, respectively. Diabetic rats were prepared by injection of streptozotocin (50 mg/kg, i.p.). The expression levels of SRC-1 and SRC-3 were significantly decreased in endothelial cells from the diabetic rats. Similar phenomenon was also observed in aortic endothelial cells from the normal rats treated with a high glucose (25 mM) for 4 h or 8 h. The expression levels of SRC-2 were little affected by hyperglycemia. Overexpression of SRC-1 and SRC-3 in high glucose-treated endothelial cells significantly increased the cell viability, suspended cell senescence, and inhibited cell apoptosis compared with the control cells. We further showed that overexpression of SRC-1 and SRC-3 markedly suppressed endothelial injury through restoring nitric oxide production, upregulating the expression of antioxidant enzymes (SOD, GPX, and CAT), and activating the PI3K/Akt pathway. The beneficial effects of SRC-1 and SRC-3 overexpression were blocked by treatment with the PI3K inhibitor LY294002 (10 mM) or with the Akt inhibitor MK-2206 (100 nM). In conclusion, hyperglycemia decreased SRC-1 and SRC-3 expression levels in rat aortic endothelial cells. SRC-1 and SRC-3 overexpression might protect against endothelial injury via inhibition of oxidative stress and activation of PI3K/Akt pathway.
Collapse
Affiliation(s)
- Xiao-Juan Quan
- Department of Geriatrics & Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| | - Chun-Lian Liang
- Department of Geriatrics & Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ming-Zhu Sun
- Department of Geriatrics & Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Lin Zhang
- Department of Geriatrics & Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Xiu-Li Li
- Department of Geriatrics & Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| |
Collapse
|
8
|
Nam DH, Kim E, Benham A, Park HK, Soibam B, Taffet GE, Kaelber JT, Suh JH, Taegtmeyer H, Entman ML, Reineke EL. Transient activation of AMPK preceding left ventricular pressure overload reduces adverse remodeling and preserves left ventricular function. FASEB J 2018; 33:711-721. [PMID: 30024790 DOI: 10.1096/fj.201800602r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Coordinated changes in signaling pathways and gene expression in hearts subjected to prolonged stress maintain cardiac function. Loss of steroid receptor coactivator-2 (SRC-2) results in a reversal to the fetal gene program and disrupts the response to pressure overload, accompanied by prominent effects on metabolism and growth signaling, including increased AMPK activation. We proposed that early metabolic stress driven by AMPK activation induces contractile dysfunction in mice lacking SRC-2. We used 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) to activate AMPK transiently before transverse aortic constriction (TAC) in wild-type and cardiomyocyte-specific SRC-2 knockout (CKO) animals. In contrast to AMPK activities during stress, in unstressed hearts, AICAR induced a mild activation of Akt signaling, and, in SRC-2-CKO mice, partially relieved an NAD+ deficiency and increased antioxidant signaling. These molecular changes translated to a mild hypertrophic response to TAC with decreased maladaptive remodeling, including markedly decreased fibrosis. Additionally, preactivation of AMPK in SRC-2-CKO mice was accompanied by a dramatic improvement in cardiac function compared with saline-treated SRC-2-CKO mice. Our results show that altered molecular signaling before stress onset has extended effects on sustained cardiac stress responses, and prestress modulation of transient growth and metabolism pathways may control those effects.-Nam, D. H., Kim, E., Benham, A., Park, H.-K., Soibam, B., Taffet, G. E., Kaelber, J. T., Suh, J. H., Taegtmeyer, H., Entman, M. L., Reineke, E. L. Transient activation of AMPK preceding left ventricular pressure overload reduces adverse remodeling and preserves left ventricular function.
Collapse
Affiliation(s)
- Deok Hwa Nam
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Eunah Kim
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | | | - Hye-Kyung Park
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Benjamin Soibam
- Department of Computer Science and Engineering Technology, University of Houston-Downtown, Houston, Texas, USA
| | - George E Taffet
- Division of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jason T Kaelber
- National Center for Macromolecular Imaging, Baylor College of Medicine, Houston, Texas, USA.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA; and
| | - Ji Ho Suh
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, USA
| | - Mark L Entman
- Division of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Erin L Reineke
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
9
|
Suh JH, Lai L, Nam D, Kim J, Jo J, Taffet GE, Kim E, Kaelber JT, Lee HK, Entman ML, Cooke JP, Reineke EL. Steroid receptor coactivator-2 (SRC-2) coordinates cardiomyocyte paracrine signaling to promote pressure overload-induced angiogenesis. J Biol Chem 2017; 292:21643-21652. [PMID: 29127200 DOI: 10.1074/jbc.m117.804740] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/09/2017] [Indexed: 12/23/2022] Open
Abstract
Pressure overload-induced cardiac stress induces left ventricular hypertrophy driven by increased cardiomyocyte mass. The increased energetic demand and cardiomyocyte size during hypertrophy necessitate increased fuel and oxygen delivery and stimulate angiogenesis in the left ventricular wall. We have previously shown that the transcriptional regulator steroid receptor coactivator-2 (SRC-2) controls activation of several key cardiac transcription factors and that SRC-2 loss results in extensive cardiac transcriptional remodeling. Pressure overload in mice lacking SRC-2 induces an abrogated hypertrophic response and decreases sustained cardiac function, but the cardiomyocyte-specific effects of SRC-2 in these changes are unknown. Here, we report that cardiomyocyte-specific loss of SRC-2 (SRC-2 CKO) results in a blunted hypertrophy accompanied by a rapid, progressive decrease in cardiac function. We found that SRC-2 CKO mice exhibit markedly decreased left ventricular vasculature in response to transverse aortic constriction, corresponding to decreased expression of the angiogenic factor VEGF. Of note, SRC-2 knockdown in cardiomyocytes decreased VEGF expression and secretion to levels sufficient to blunt in vitro tube formation and proliferation of endothelial cells. During pressure overload, both hypertrophic and hypoxic signals can stimulate angiogenesis, both of which stimulated SRC-2 expression in vitro Furthermore, SRC-2 coactivated the transcription factors GATA-binding protein 4 (GATA-4) and hypoxia-inducible factor (HIF)-1α and -2α in response to angiotensin II and hypoxia, respectively, which drive VEGF expression. These results suggest that SRC-2 coordinates cardiomyocyte secretion of VEGF downstream of the two major angiogenic stimuli occurring during pressure overload bridging both hypertrophic and hypoxia-stimulated paracrine signaling.
Collapse
Affiliation(s)
- Ji Ho Suh
- From the Center for Bioenergetics and
| | - Li Lai
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas 77030
| | | | - Jong Kim
- the University of Houston 77004, Houston, Texas
| | - Juyeon Jo
- the Department of Pediatrics and Neuroscience, Baylor College of Medicine and Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030, and
| | - George E Taffet
- the Division of Cardiovascular Sciences, Department of Medicine, and
| | - Eunah Kim
- From the Center for Bioenergetics and
| | - Jason T Kaelber
- the National Center for Macromolecular Imaging and Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030
| | - Hyun-Kyoung Lee
- the Department of Pediatrics and Neuroscience, Baylor College of Medicine and Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030, and
| | - Mark L Entman
- the Division of Cardiovascular Sciences, Department of Medicine, and
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas 77030
| | | |
Collapse
|
10
|
Nam D, Reineke EL. Timing and Targeting of Treatment in Left Ventricular Hypertrophy. Methodist Debakey Cardiovasc J 2017; 13:9-14. [PMID: 28413576 DOI: 10.14797/mdcj-13-1-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
In most clinical cases, left ventricular hypertrophy (LVH) occurs over time from persistent cardiac stress. At the molecular level, this results in both transient and long-term changes to metabolic, sarcomeric, ion handling, and stress signaling pathways. Although this is initially an adaptive change, the mechanisms underlying LVH eventually lead to maladaptive changes including fibrosis, decreased cardiac function, and failure. Understanding the regulators of long-term changes, which are largely driven by transcriptional remodeling, is a crucial step in identifying novel therapeutic targets for preventing the downstream negative effects of LVH and treatments that could reverse or prevent it. The development of effective therapeutics, however, will require a critical understanding of what to target, how to modify important pathways, and how to identify the stage of pathology in which a specific treatment should be used.
Collapse
Affiliation(s)
- Deokhwa Nam
- Houston Methodist Research Institute, Houston, Texas
| | | |
Collapse
|
11
|
Fleet T, Zhang B, Lin F, Zhu B, Dasgupta S, Stashi E, Tackett B, Thevananther S, Rajapakshe KI, Gonzales N, Dean A, Mao J, Timchenko N, Malovannaya A, Qin J, Coarfa C, DeMayo F, Dacso CC, Foulds CE, O'Malley BW, York B. SRC-2 orchestrates polygenic inputs for fine-tuning glucose homeostasis. Proc Natl Acad Sci U S A 2015; 112:E6068-77. [PMID: 26487680 PMCID: PMC4640775 DOI: 10.1073/pnas.1519073112] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Despite extensive efforts to understand the monogenic contributions to perturbed glucose homeostasis, the complexity of genetic events that fractionally contribute to the spectrum of this pathology remain poorly understood. Proper maintenance of glucose homeostasis is the central feature of a constellation of comorbidities that define the metabolic syndrome. The ability of the liver to balance carbohydrate uptake and release during the feeding-to-fasting transition is essential to the regulation of peripheral glucose availability. The liver coordinates the expression of gene programs that control glucose absorption, storage, and secretion. Herein, we demonstrate that Steroid Receptor Coactivator 2 (SRC-2) orchestrates a hierarchy of nutritionally responsive transcriptional complexes to precisely modulate plasma glucose availability. Using DNA pull-down technology coupled with mass spectrometry, we have identified SRC-2 as an indispensable integrator of transcriptional complexes that control the rate-limiting steps of hepatic glucose release and accretion. Collectively, these findings position SRC-2 as a major regulator of polygenic inputs to metabolic gene regulation and perhaps identify a previously unappreciated model that helps to explain the clinical spectrum of glucose dysregulation.
Collapse
Affiliation(s)
- Tiffany Fleet
- Interdepartmental Department in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030-3411; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Bin Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Fumin Lin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Bokai Zhu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Subhamoy Dasgupta
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Erin Stashi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Bryan Tackett
- Department of Pediatrics, Gastroenterology, Hepatology & Nutrition, Baylor College of Medicine, Houston, TX 77030-3411
| | - Sundararajah Thevananther
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411; Department of Pediatrics, Gastroenterology, Hepatology & Nutrition, Baylor College of Medicine, Houston, TX 77030-3411
| | - Kimal I Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Naomi Gonzales
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Adam Dean
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Jianqiang Mao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Nikolai Timchenko
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3026
| | - Anna Malovannaya
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Jun Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Francesco DeMayo
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030-3411
| | - Clifford C Dacso
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411; Department of Medicine, Baylor College of Medicine, Houston, TX 77030-3411
| | - Charles E Foulds
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411
| | - Bert W O'Malley
- Interdepartmental Department in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030-3411; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030-3411;
| | - Brian York
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030-3411; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030-3411;
| |
Collapse
|
12
|
Qin J, Lee HJ, Wu SP, Lin SC, Lanz RB, Creighton CJ, DeMayo FJ, Tsai SY, Tsai MJ. Androgen deprivation-induced NCoA2 promotes metastatic and castration-resistant prostate cancer. J Clin Invest 2014; 124:5013-26. [PMID: 25295534 DOI: 10.1172/jci76412] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 09/04/2014] [Indexed: 01/05/2023] Open
Abstract
A major clinical hurdle for the management of advanced prostate cancer (PCa) in patients is the resistance of tumors to androgen deprivation therapy (ADT) and their subsequent development into castration-resistant prostate cancer (CRPC). While recent studies have identified potential pathways involved in CRPC development, the drivers of CRPC remain largely undefined. Here we determined that nuclear receptor coactivator 2 (NCoA2, also known as SRC-2), which is frequently amplified or overexpressed in patients with metastatic PCa, mediates development of CRPC. In a murine model, overexpression of NCoA2 in the prostate epithelium resulted in neoplasia and, in combination with Pten deletion, promoted the development of metastasis-prone cancer. Moreover, depletion of NCoA2 in PTEN-deficient mice prevented the development of CRPC. In human androgen-sensitive prostate cancer cells, androgen signaling suppressed NCoA2 expression, and NCoA2 overexpression in murine prostate tumors resulted in hyperactivation of PI3K/AKT and MAPK signaling, promoting tumor malignance. Analysis of PCa patient samples revealed a strong correlation among NCoA2-mediated signaling, disease progression, and PCa recurrence. Taken together, our findings indicate that androgen deprivation induces NCoA2, which in turn mediates activation of PI3K signaling and promotes PCa metastasis and CRPC development. Moreover, these results suggest that the inhibition of NCoA2 has potential for PCa therapy.
Collapse
|
13
|
Dasgupta S, O'Malley BW. Transcriptional coregulators: emerging roles of SRC family of coactivators in disease pathology. J Mol Endocrinol 2014; 53:R47-59. [PMID: 25024406 PMCID: PMC4152414 DOI: 10.1530/jme-14-0080] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Transcriptional coactivators have evolved as an important new class of functional proteins that participate with virtually all transcription factors and nuclear receptors (NRs) to intricately regulate gene expression in response to a wide variety of environmental cues. Recent findings have highlighted that coactivators are important for almost all biological functions, and consequently, genetic defects can lead to severe pathologies. Drug discovery efforts targeting coactivators may prove valuable for treatment of a variety of diseases.
Collapse
Affiliation(s)
- Subhamoy Dasgupta
- Department of Molecular and Cellular BiologyBaylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular BiologyBaylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| |
Collapse
|
14
|
Stashi E, York B, O'Malley BW. Steroid receptor coactivators: servants and masters for control of systems metabolism. Trends Endocrinol Metab 2014; 25:337-47. [PMID: 24953190 PMCID: PMC4108168 DOI: 10.1016/j.tem.2014.05.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 11/30/2022]
Abstract
Coregulator recruitment to nuclear receptors (NRs) and other transcription factors is essential for proper metabolic gene regulation, with coactivators enhancing and corepressors attenuating gene transcription. The steroid receptor coactivator (SRC) family is composed of three homologous members (SRC-1, SRC-2, and SRC-3), which are uniquely important for mediating steroid hormone and mitogenic actions. An accumulating body of work highlights the diverse array of metabolic functions regulated by the SRCs, including systemic metabolite homeostasis, inflammation, and energy regulation. We discuss here the cooperative and unique functions among the SRCs to provide a comprehensive atlas of systemic SRC metabolic regulation. Deciphering the fractional and synergistic contributions of the SRCs to metabolic homeostasis is crucial to understanding fully the networks underlying metabolic transcriptional regulation.
Collapse
Affiliation(s)
- Erin Stashi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Brian York
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
15
|
Szwarc MM, Kommagani R, Jeong JW, Wu SP, Tsai SY, Tsai MJ, O’Malley BW, DeMayo FJ, Lydon JP. Perturbing the cellular levels of steroid receptor coactivator-2 impairs murine endometrial function. PLoS One 2014; 9:e98664. [PMID: 24905738 PMCID: PMC4048228 DOI: 10.1371/journal.pone.0098664] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/01/2014] [Indexed: 01/24/2023] Open
Abstract
As pleiotropic coregulators, members of the p160/steroid receptor coactivator (SRC) family control a broad spectrum of transcriptional responses that underpin a diverse array of physiological and pathophysiological processes. Because of their potent coregulator properties, strict controls on SRC expression levels are required to maintain normal tissue functionality. Accordingly, an unwarranted increase in the cellular levels of SRC members has been causally linked to the initiation and/or progression of a number of clinical disorders. Although knockout mouse models have underscored the critical non-redundant roles for each SRC member in vivo, there are surprisingly few mouse models that have been engineered to overexpress SRCs. This deficiency is significant since SRC involvement in many of these disorders is based on unscheduled increases in the levels (rather than the absence) of SRC expression. To address this deficiency, we used recent mouse technology that allows for the targeted expression of human SRC-2 in cells which express the progesterone receptor. Through cre-loxP recombination driven by the endogenous progesterone receptor promoter, a marked elevation in expression levels of human SRC-2 was achieved in endometrial cells that are positive for the progesterone receptor. As a result of this increase in coregulator expression, female mice are severely subfertile due to a dysfunctional uterus, which exhibits a hypersensitivity to estrogen exposure. Our findings strongly support the proposal from clinical observations that increased levels of SRC-2 are causal for a number of endometrial disorders which compromise fertility. Future studies will use this mouse model to decipher the molecular mechanisms that underpin the endometrial defect. We believe such mechanistic insight may provide new molecular descriptors for diagnosis, prognosis, and/or therapy in the clinical management of female infertility.
Collapse
Affiliation(s)
- Maria M. Szwarc
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ramakrishna Kommagani
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, Michigan, United States of America
| | - San-Pin Wu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sophia Y. Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ming-Jer Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Bert W. O’Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Francesco J. DeMayo
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
16
|
Eraly SA. Striking differences between knockout and wild-type mice in global gene expression variability. PLoS One 2014; 9:e97734. [PMID: 24830645 PMCID: PMC4022672 DOI: 10.1371/journal.pone.0097734] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 04/23/2014] [Indexed: 12/16/2022] Open
Abstract
Microarray analyses of gene knockouts have traditionally focused on the identification of genes whose mean expression is different in knockout and wild-type mice. However, recent work suggests that changes in the variability of gene expression can have important phenotypic consequences as well. Here, in an unbiased sample of publicly available microarray data on gene expression in various knockouts, highly significant differences from wild-type (either increases or decreases) are noted in the gene expression coefficients of variation (CVs) of virtually every knockout considered. Examination of the distribution of gene-by-gene CV differences indicates that these findings are not attributable to a few outlier genes, but rather to broadly increased or decreased CV in the various knockouts over all the (tens of thousands of) transcripts assayed. These global differences in variability may reflect either authentic biological effects of the knockouts or merely experimental inconsistencies. However, regardless of the underlying explanation, the variability differences are of importance as they will influence both the statistical detection of gene expression changes and, potentially, the knockout phenotype itself.
Collapse
Affiliation(s)
- Satish A. Eraly
- Department of Medicine, University of California San Diego, School of Medicine, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
17
|
Reineke EL, Benham A, Soibam B, Stashi E, Taegtmeyer H, Entman ML, Schwartz RJ, O'Malley BW. Steroid receptor coactivator-2 is a dual regulator of cardiac transcription factor function. J Biol Chem 2014; 289:17721-31. [PMID: 24811170 DOI: 10.1074/jbc.m113.539908] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
We have previously demonstrated the potential role of steroid receptor coactivator-2 (SRC-2) as a co-regulator in the transcription of critical molecules modulating cardiac function and metabolism in normal and stressed hearts. The present study seeks to extend the previous information by demonstrating SRC-2 fulfills this role by serving as a critical coactivator for the transcription and activity of critical transcription factors known to control cardiac growth and metabolism as well as in their downstream signaling. This knowledge broadens our understanding of the mechanism by which SRC-2 acts in normal and stressed hearts and allows further investigation of the transcriptional modifications mediating different types and degrees of cardiac stress. Moreover, the genetic manipulation of SRC-2 in this study is specific for the heart and thereby eliminating potential indirect effects of SRC-2 deletion in other organs. We have shown that SRC-2 is critical to transcriptional control modulated by MEF2, GATA-4, and Tbx5, thereby enhancing gene expression associated with cardiac growth. Additionally, we describe SRC-2 as a novel regulator of PPARα expression, thus controlling critical steps in metabolic gene expression. We conclude that through regulation of cardiac transcription factor expression and activity, SRC-2 is a critical transcriptional regulator of genes important for cardiac growth, structure, and metabolism, three of the main pathways altered during the cardiac stress response.
Collapse
Affiliation(s)
- Erin L Reineke
- From the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Ashley Benham
- the Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas 77030
| | - Benjamin Soibam
- the Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas 77030
| | - Erin Stashi
- From the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Heinrich Taegtmeyer
- the Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, Houston, Texas 77030
| | - Mark L Entman
- the Department of Medicine, Division of Cardiovascular Sciences, Baylor College of Medicine, Houston, Texas 77030, and
| | - Robert J Schwartz
- the Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas 77030, the Department of Biology and Biochemistry, University of Houston, Houston, Texas 77004
| | - Bert W O'Malley
- From the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030,
| |
Collapse
|
18
|
Wang S, Gong H, Jiang G, Ye Y, Wu J, You J, Zhang G, Sun A, Komuro I, Ge J, Zou Y. Src is required for mechanical stretch-induced cardiomyocyte hypertrophy through angiotensin II type 1 receptor-dependent β-arrestin2 pathways. PLoS One 2014; 9:e92926. [PMID: 24699426 PMCID: PMC3974699 DOI: 10.1371/journal.pone.0092926] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 02/26/2014] [Indexed: 01/14/2023] Open
Abstract
Angiotensin II (AngII) type 1 receptor (AT1-R) can be activated by mechanical stress (MS) without the involvement of AngII during the development of cardiomyocyte hypertrophy, in which G protein-independent pathways are critically involved. Although β-arrestin2-biased signaling has been speculated, little is known about how AT1-R/β-arrestin2 leads to ERK1/2 activation. Here, we present a novel mechanism by which Src kinase mediates AT1-R/β-arrestin2-dependent ERK1/2 phosphorylation in response to MS. Differing from stimulation by AngII, MS-triggered ERK1/2 phosphorylation is neither suppressed by overexpression of RGS4 (the negative regulator of the G-protein coupling signal) nor by inhibition of Gαq downstream protein kinase C (PKC) with GF109203X. The release of inositol 1,4,5-triphosphate (IP3) is increased by AngII but not by MS. These results collectively suggest that MS-induced ERK1/2 activation through AT1-R might be independent of G-protein coupling. Moreover, either knockdown of β-arrestin2 or overexpression of a dominant negative mutant of β-arrestin2 prevents MS-induced activation of ERK1/2. We further identifies a relationship between Src, a non-receptor tyrosine kinase and β-arrestin2 using analyses of co-immunoprecipitation and immunofluorescence after MS stimulation. Furthermore, MS-, but not AngII-induced ERK1/2 phosphorylation is attenuated by Src inhibition, which also significantly improves pressure overload-induced cardiac hypertrophy and dysfunction in mice lacking AngII. Finally, MS-induced Src activation and hypertrophic response are abolished by candesartan but not by valsartan whereas AngII-induced responses can be abrogated by both blockers. Our results suggest that Src plays a critical role in MS-induced cardiomyocyte hypertrophy through β-arrestin2-associated angiotensin II type 1 receptor signaling.
Collapse
MESH Headings
- Angiotensinogen/physiology
- Animals
- Animals, Newborn
- Arrestins/genetics
- Arrestins/metabolism
- Blotting, Western
- Cardiomegaly/metabolism
- Cardiomegaly/pathology
- Cells, Cultured
- Echocardiography
- Immunoenzyme Techniques
- Immunoprecipitation
- Inositol 1,4,5-Trisphosphate/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phosphorylation
- RNA, Messenger/genetics
- Rats
- Real-Time Polymerase Chain Reaction
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Stress, Mechanical
- beta-Arrestins
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Guoliang Jiang
- Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Yong Ye
- Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Jian Wu
- Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Jieyun You
- Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Guoping Zhang
- Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Aijun Sun
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Issei Komuro
- Department of Cardiovascular Medicine, the University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institutes of Biomedical Science, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Motamed M, Rajapakshe KI, Hartig SM, Coarfa C, Moses RE, Lonard DM, O'Malley BW. Steroid receptor coactivator 1 is an integrator of glucose and NAD+/NADH homeostasis. Mol Endocrinol 2014; 28:395-405. [PMID: 24438340 DOI: 10.1210/me.2013-1404] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Steroid receptor coactivator 1 (SRC-1) drives diverse gene expression programs necessary for the dynamic regulation of cancer metastasis, inflammation and gluconeogenesis, pointing to its overlapping roles as an oncoprotein and integrator of cell metabolic programs. Nutrient utilization has been intensely studied with regard to cellular adaptation in both cancer and noncancerous cells. Nonproliferating cells consume glucose through the citric acid cycle to generate NADH to fuel ATP generation via mitochondrial oxidative phosphorylation. In contrast, cancer cells undergo metabolic reprogramming to support rapid proliferation. To generate lipids, nucleotides, and proteins necessary for cell division, most tumors switch from oxidative phosphorylation to glycolysis, a phenomenon known as the Warburg Effect. Because SRC-1 is a key coactivator responsible for driving a hepatic gluconeogenic program under fasting conditions, we asked whether SRC-1 responds to alterations in nutrient availability to allow for adaptive metabolism. Here we show SRC-1 is stabilized by the 26S proteasome in the absence of glucose. RNA profiling was used to examine the effects of SRC-1 perturbation on gene expression in the absence or presence of glucose, revealing that SRC-1 affects the expression of complex I of the mitochondrial electron transport chain, a set of enzymes responsible for the conversion of NADH to NAD(+). NAD(+) and NADH were subsequently identified as metabolites that underlie SRC-1's response to glucose deprivation. Knockdown of SRC-1 in glycolytic cancer cells abrogated their ability to grow in the absence of glucose consistent with SRC-1's role in promoting cellular adaptation to reduced glucose availability.
Collapse
Affiliation(s)
- Massoud Motamed
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | | | | | | | | | | | | |
Collapse
|
20
|
Dasgupta S, Lonard DM, O'Malley BW. Nuclear receptor coactivators: master regulators of human health and disease. Annu Rev Med 2013; 65:279-92. [PMID: 24111892 DOI: 10.1146/annurev-med-051812-145316] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Transcriptional coregulators (coactivators and corepressors) have emerged as the principal modulators of the functions of nuclear receptors and other transcription factors. During the decade since the discovery of steroid receptor coactivator-1 (SRC-1), the first authentic coregulator, more than 400 coregulators have been identified and characterized, and deciphering their function has contributed significantly to our understanding of their role in human physiology. Deregulated expression of coregulators has been implicated in diverse disease states and related pathologies. The advancement of molecular technologies has enabled us to better characterize the molecular associations of the SRC family of coactivators with other protein complexes in the context of gene regulation. These continuing discoveries not only expand our knowledge of the roles of coactivators in various human diseases but allow us to discover novel coactivator-targeting strategies for therapeutic intervention in these diseases.
Collapse
Affiliation(s)
- Subhamoy Dasgupta
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030;
| | | | | |
Collapse
|
21
|
Fenne IS, Helland T, Flågeng MH, Dankel SN, Mellgren G, Sagen JV. Downregulation of steroid receptor coactivator-2 modulates estrogen-responsive genes and stimulates proliferation of mcf-7 breast cancer cells. PLoS One 2013; 8:e70096. [PMID: 23936147 PMCID: PMC3728357 DOI: 10.1371/journal.pone.0070096] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/14/2013] [Indexed: 11/18/2022] Open
Abstract
The p160/Steroid Receptor Coactivators SRC-1, SRC-2/GRIP1, and SRC-3/AIB1 are important regulators of Estrogen Receptor alpha (ERα) activity. However, whereas the functions of SRC-1 and SRC-3 in breast tumourigenesis have been extensively studied, little is known about the role of SRC-2. Previously, we reported that activation of the cAMP-dependent protein kinase, PKA, facilitates ubiquitination and proteasomal degradation of SRC-2 which in turn leads to inhibition of SRC-2-coactivation of ERα and changed expression of the ERα target gene, pS2. Here we have characterized the global program of transcription in SRC-2-depleted MCF-7 breast cancer cells using short-hairpin RNA technology, and in MCF-7 cells exposed to PKA activating agents. In order to identify genes that may be regulated through PKA-induced downregulation of SRC-2, overlapping transcriptional targets in response to the respective treatments were characterized. Interestingly, we observed decreased expression of several breast cancer tumour suppressor genes (e.g., TAGLN, EGR1, BCL11b, CAV1) in response to both SRC-2 knockdown and PKA activation, whereas the expression of a number of other genes implicated in cancer progression (e.g., RET, BCAS1, TFF3, CXCR4, ADM) was increased. In line with this, knockdown of SRC-2 also stimulated proliferation of MCF-7 cells. Together, these results suggest that SRC-2 may have an antiproliferative function in breast cancer cells.
Collapse
Affiliation(s)
- Ingvild S. Fenne
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Thomas Helland
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Marianne H. Flågeng
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Simon N. Dankel
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Jørn V. Sagen
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
22
|
Zhou L, Chen Z, Vanderslice P, So SP, Ruan KH, Willerson JT, Dixon RAF. Endothelial-like progenitor cells engineered to produce prostacyclin rescue monocrotaline-induced pulmonary arterial hypertension and provide right ventricle benefits. Circulation 2013; 128:982-94. [PMID: 23841984 DOI: 10.1161/circulationaha.113.003139] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Intravenous prostacyclin is approved for treating pulmonary arterial hypertension (PAH), but it has a short half-life and must be delivered systemically via an indwelling intravenous catheter. We hypothesize that localized jugular vein delivery of prostacyclin-producing cells may provide sustained therapeutic effects without the limitations of systemic delivery. METHODS AND RESULTS We generated a vector expressing a human cyclooxygenase isoform 1 and prostacyclin synthase fusion protein that produces prostacyclin from arachidonic acid. Endothelial-like progenitor cells (ELPCs) were transfected with the cyclooxygenase isoform 1-prostacyclin synthase plasmid and labeled with lentivirus expressing nuclear-localized red fluorescent protein (nuRFP). The engineered ELPCs (expressing cyclooxygenase isoform 1-prostacyclin synthase and nuRFP) were tested in rats with monocrotaline (MCT)-induced PAH. In PAH prevention studies, treatment with engineered ELPCs or control ELPCs (expressing nuRFP alone) attenuated MCT-induced right ventricular systolic pressure increase, right ventricular hypertrophy, and pulmonary vessel wall thickening. Engineered ELPCs were more effective than control ELPCs in all variables evaluated. In PAH reversal studies, engineered ELPCs or control ELPCs increased the survival rate of rats with established PAH and decreased right ventricular hypertrophy. Engineered ELPCs provided a survival benefit 2 weeks earlier than did control ELPCs. Microarray-based gene ontology analysis of the right ventricle revealed that a number of MCT-altered genes and neurotransmitter pathways (dopamine, serotonin, and γ-aminobutyric acid) were restored after ELPC-based prostacyclin gene therapy. CONCLUSIONS Cyclooxygenase isoform 1-prostacyclin synthase-expressing ELPCs reversed MCT-induced PAH. A single jugular vein injection offered survival benefits for at least 4 weeks and may provide a promising option for PAH patients.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Molecular Cardiology, Texas Heart Institute, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|