51
|
Nawroth J, Rogal J, Weiss M, Brucker SY, Loskill P. Organ-on-a-Chip Systems for Women's Health Applications. Adv Healthc Mater 2018; 7. [PMID: 28985032 DOI: 10.1002/adhm.201700550] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/30/2017] [Indexed: 12/19/2022]
Abstract
Biomedical research, for a long time, has paid little attention to the influence of sex in many areas of study, ranging from molecular and cellular biology to animal models and clinical studies on human subjects. Many studies solely rely on male cells/tissues/animals/humans, although there are profound differences in male and female physiology, which can significantly impact disease mechanisms, toxicity of compounds, and efficacy of pharmaceuticals. In vitro systems have been traditionally very limited in their capacity to recapitulate female-specific physiology and anatomy such as dynamic sex-hormone levels and the complex interdependencies of female reproductive tract organs. However, the advent of microphysiological organ-on-a-chip systems, which attempt to recreate the 3D structure and function of human organs, now gives researchers the opportunity to integrate cells and tissues from a variety of individuals. Moreover, adding a dynamic flow environment allows mimicking endocrine signaling during the menstrual cycle and pregnancy, as well as providing a controlled microfluidic environment for pharmacokinetic modeling. This review gives an introduction into preclinical and clinical research on women's health and discusses where organ-on-a-chip systems are already utilized or have the potential to deliver new insights and enable entirely new types of studies.
Collapse
Affiliation(s)
| | - Julia Rogal
- Department of Cell and Tissue Engineering; Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB; Nobelstrasse 12 70569 Stuttgart Germany
| | - Martin Weiss
- Department of Gynecology and Obstetrics; University Medicine Tübingen; Calwerstrasse 7 72076 Tübingen Germany
| | - Sara Y. Brucker
- Department of Gynecology and Obstetrics; University Medicine Tübingen; Calwerstrasse 7 72076 Tübingen Germany
| | - Peter Loskill
- Department of Cell and Tissue Engineering; Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB; Nobelstrasse 12 70569 Stuttgart Germany
| |
Collapse
|
52
|
Effects of Female Sex Steroids Administration on Pathophysiologic Mechanisms in Traumatic Brain Injury. Transl Stroke Res 2017; 9:393-416. [PMID: 29151229 DOI: 10.1007/s12975-017-0588-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/16/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
Secondary brain damage following initial brain damage in traumatic brain injury (TBI) is a major cause of adverse outcomes. There are many gaps in TBI research and a lack of therapy to limit debilitating outcomes in TBI or enhance the neurogenesis, despite pre-clinical and clinical research performed in TBI. Females show harmful outcomes against brain damage including TBI less than males, independent of different TBI occurrence. A significant reduction in secondary brain damage and improvement in neurologic outcome post-TBI has been reported following the use of progesterone and estrogen in many experimental studies. Although useful features of sex steroids including progesterone have been identified in TBI clinical trials I and II, clinical trials III have been unsuccessful. This review article focuses on evidence of secondary injury mechanisms and neuroprotective effects of estrogen and progesterone in TBI. Understanding these mechanisms may enable researchers to achieve greater success in TBI clinical studies. It seems that the design of clinical studies should be revised due to translation loss of animal studies to clinical studies. The heterogeneous and complex nature of TBI, the endogenous levels of sex hormones at the time of taking these hormones, the therapeutic window of the drug, the dosage of the drug, the selection of appropriate targets in evaluation, the determination of responsive population, gender and age based on animal studies should be considered in the design of TBI human studies in future.
Collapse
|
53
|
Zheng Y, Liu L, Chen C, Ming P, Huang Q, Li C, Cao D, Xu X, Ge W. The extracellular vesicles secreted by lung cancer cells in radiation therapy promote endothelial cell angiogenesis by transferring miR-23a. PeerJ 2017; 5:e3627. [PMID: 28852584 PMCID: PMC5572936 DOI: 10.7717/peerj.3627] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 07/10/2017] [Indexed: 01/01/2023] Open
Abstract
Angiogenesis is an important factor contributing to the radioresistance of lung cancer. However, the associated mechanisms underlying radiotherapy-induced pro-angiogenesis are unclear. Here, we demonstrated that Extracellular vesicles (EVs) derived from cultured cells in vitro enhanced HUVEC proliferation and migration, and the enhancement effect became more obvious when HUVECs were treated with EV derived from A549 or H1299, two lung cancer cell lines. Additionally, the pro-angiogenesis effect induced by EV could be strengthened when the lung cancer cells were exposed to X-ray irradiation. Furthermore, we verified that the downregulation of PTEN plays a vital role in this process. By evaluating the changes in the levels of microRNAs(miRNAs) targeting PTEN in EV, we found that miR-23a was significantly upregulated and mediated a decrease in PTEN. A luciferase reporter gene transfer experiment demonstrated that PTEN was the direct target of miR-23a, and the kinetics of PTEN expression were opposite to those of miR-23a. Our results show that the miR-23a/PTEN pathway plays an important role in EV-induced angiogenesis. These findings implicate the miR-23a/PTEN axis as a novel therapeutic target for lung cancer radiotherapy.
Collapse
Affiliation(s)
- Yongfa Zheng
- Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Liang Liu
- Shanghai Cancer Center, Fudan University, Shanghai, Shanghai, China
| | - Cong Chen
- Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Pingpo Ming
- Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qin Huang
- Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Changhu Li
- Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Dedong Cao
- Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Ximing Xu
- Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Wei Ge
- Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
54
|
Eldien MMS, Abdou AG, Rageh T, Abdelrazek E, Elkholy E. Immunohistochemical expression of ER-α and PR in papillary thyroid carcinoma. Ecancermedicalscience 2017; 11:748. [PMID: 28717394 PMCID: PMC5493440 DOI: 10.3332/ecancer.2017.748] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Indexed: 11/06/2022] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common thyroid cancer with multiple risk factors including exposure to ionising radiation. Oestrogens contribute to papillary carcinoma development by promoting cell proliferation and invasion of mutated epithelial follicular cells. The present study aimed to assess ER-α and PR expression in PTC and to correlate their expression with the clinicopathological parameters in this cancer. This study included 62 primary and six metastatic papillary thyroid carcinoma cases. Nineteen and 38.7% of primary PTC cases showed positive nuclear expression for ER and PR, respectively. Metastatic cases showed 66.7% positive ER expression and all were negative for PR. Oestrogen receptor expression showed significant higher positivity in metastatic compared to primary PTC (p = 0.02) and it was significantly associated with primary PTC associated with thyroiditis (p = .002). Progesterone receptor expression was significantly associated with old age in primary PTC (p = .003) and it showed significant coparallel expression with ER (p = .000). Oestrogen and progesterone receptors expressed in papillary thyroid carcinoma opening the door for further studies to verify if those patients could benefit from hormonal therapy. Oestrogen receptor seems to have a role in metastatic process of PTC as malignant cells express it in metastatic more than primary site. The presence of lymphocytes in the stroma may promote ER expression in adjacent PTC, necessitating further studies on PTC cases associated with Hashimoto thyroiditis to verify this assumed relationship.
Collapse
Affiliation(s)
- Marwa Mohammed Serag Eldien
- Department of Pathology, Faculty of Medicine, Menoufia University, Gamal Abd-Elnaser street, Shebein Elkom 32511, Egypt
| | - Asmaa Gaber Abdou
- Department of Pathology, Faculty of Medicine, Menoufia University, Gamal Abd-Elnaser street, Shebein Elkom 32511, Egypt
| | - Tarek Rageh
- Department of Surgery, Faculty of Medicine, Menoufia University, Gamal Abd-Elnaser street, Shebein Elkom 32511, Egypt
| | - Eman Abdelrazek
- Department of Oncology, Faculty of Medicine, Menoufia University, Gamal Abd-Elnaser street, Shebein Elkom 32511, Egypt
| | - Enas Elkholy
- Department of Oncology, Faculty of Medicine, Menoufia University, Gamal Abd-Elnaser street, Shebein Elkom 32511, Egypt
| |
Collapse
|
55
|
Landeros RV, Jobe SO, Aranda-Pino G, Lopez GE, Zheng J, Magness RR. Convergent ERK1/2, p38 and JNK mitogen activated protein kinases (MAPKs) signalling mediate catecholoestradiol-induced proliferation of ovine uterine artery endothelial cells. J Physiol 2017; 595:4663-4676. [PMID: 28437005 DOI: 10.1113/jp274119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/10/2017] [Indexed: 12/25/2022] Open
Abstract
KEY POINTS The catechol metabolites of 17β-oestradiol (E2 β), 2-hydroxyoestradiol (2-OHE2 ) and 4-hydroxyoestradiol (4-OHE2 ), stimulate proliferation of pregnancy-derived ovine uterine artery endothelial cells (P-UAECs) through β-adrenoceptors (β-ARs) and independently of the classic oestrogen receptors (ERs). Herein we show that activation of ERK1/2, p38 and JNK mitogen activated protein kinases (MAPKs) is necessary for 2-OHE2 - and 4-OHE2 -induced P-UAEC proliferation, as well as proliferation induced by the parent hormone E2 β and other β-AR signalling hormones (i.e. catecholamines). Conversely, although 2-OHE2 and 4-OHE2 rapidly activate phosphatidylinositol 3-kinase (PI3K), its activation is not involved in catecholoestradiol-induced P-UAEC proliferation. We also show for the first time the signalling mechanisms involved in catecholoestradiol-induced P-UAEC proliferation; which converge at the level of MAPKs with the signalling mechanisms mediating E2 β- and catecholamine-induced proliferation. The present study advances our understanding of the complex signalling mechanisms involved in regulating uterine endothelial cell proliferation during pregnancy. ABSTRACT Previously we demonstrated that the biologically active metabolites of 17β-oestradiol, 2-hydroxyoestradiol (2-OHE2 ) and 4-hydroxyoestradiol (4-OHE2 ), stimulate pregnancy-specific proliferation of uterine artery endothelial cells derived from pregnant (P-UAECs), but not non-pregnant ewes. However, unlike 17β-oestradiol, which induces proliferation via oestrogen receptor-β (ER-β), the catecholoestradiols mediate P-UAEC proliferation via β-adrenoceptors (β-AR) and independently of classic oestrogen receptors. Herein, we aim to further elucidate the signalling mechanisms involved in proliferation induced by catecholoestradiols in P-UAECs. P-UAECs were treated with 2-OHE2 and 4-OHE2 for 0, 0.25, 0.5, 1, 2, 4, 12 and 24 h, to analyse activation of mitogen activated protein kinases (MAPKs) and phosphatidylinositol 3-kinase (PI3K)-AKT. Specific inhibitors for ERK1/2 MAPK (PD98059), p38 MAPK (SB203580), JNK MAPK (SP600125), or PI3K (LY294002) were used to determine the involvement of individual kinases in agonist-induced P-UAEC proliferation. 2-OHE2 and 4-OHE2 stimulated biphasic phosphorylation of ERK1/2, slow p38 and JNK phosphorylation over time, and rapid monophasic AKT phosphorylation. Furthermore, ERK1/2, p38 and JNK MAPKs, but not PI3K, were individually necessary for catecholoestradiol-induced proliferation. In addition, when comparing the signalling mechanisms of the catecholoestradiols, to 17β-oestradiol and catecholamines, we observed that convergent MAPKs signalling pathways facilitate P-UAEC proliferation induced by all of these hormones. Thus, all three members of the MAPK family mediate the mitogenic effects of catecholoestradiols in the endothelium during pregnancy. Furthermore, the convergent signalling of MAPKs involved in catecholoestradiol-, 17β-oestradiol- and catecholamine-induced endothelial cell proliferation may be indicative of unappreciated evolutionary functional redundancy to facilitate angiogenesis and ensure maintenance of uterine blood flow during pregnancy.
Collapse
Affiliation(s)
- Rosalina Villalon Landeros
- Department of Obstetrics and Gynaecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI, USA
| | - Sheikh O Jobe
- Department of Obstetrics and Gynaecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI, USA
| | - Gabrielle Aranda-Pino
- Department of Obstetrics and Gynaecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI, USA
| | - Gladys E Lopez
- Department of Obstetrics and Gynaecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI, USA
| | - Jing Zheng
- Department of Obstetrics and Gynaecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI, USA
| | - Ronald R Magness
- Department of Obstetrics and Gynaecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI, USA.,Department of Pediatrics and Animal Sciences, University of Wisconsin-Madison, Madison, WI, USA.,Department of Animal Sciences, University of Wisconsin-Madison, Madison, WI, USA.,Department of Obstetrics and Gynaecology, University of South Florida Perinatal Research Vascular Centre, Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
56
|
Berkane N, Liere P, Oudinet JP, Hertig A, Lefèvre G, Pluchino N, Schumacher M, Chabbert-Buffet N. From Pregnancy to Preeclampsia: A Key Role for Estrogens. Endocr Rev 2017; 38:123-144. [PMID: 28323944 DOI: 10.1210/er.2016-1065] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 02/28/2017] [Indexed: 02/08/2023]
Abstract
Preeclampsia (PE) results in placental dysfunction and is one of the primary causes of maternal and fetal mortality and morbidity. During pregnancy, estrogen is produced primarily in the placenta by conversion of androgen precursors originating from maternal and fetal adrenal glands. These processes lead to increased plasma estrogen concentrations compared with levels in nonpregnant women. Aberrant production of estrogens could play a key role in PE symptoms because they are exclusively produced by the placenta and they promote angiogenesis and vasodilation. Previous assessments of estrogen synthesis during PE yielded conflicting results, possibly because of the lack of specificity of the assays. However, with the introduction of reliable analytical protocols using liquid chromatography/mass spectrometry or gas chromatography/mass spectrometry, more recent studies suggest a marked decrease in estradiol levels in PE. The aim of this review is to summarize current knowledge of estrogen synthesis, regulation in the placenta, and biological effects during pregnancy and PE. Moreover, this review highlights the links among the occurrence of PE, estrogen biosynthesis, angiogenic factors, and cardiovascular risk factors. A close link between estrogen dysregulation and PE occurrence might validate estrogen levels as a biomarker but could also reveal a potential approach for prevention or cure of PE.
Collapse
Affiliation(s)
- Nadia Berkane
- Department of Gynecology and Obstetrics of University Hospital of Geneva, 1205, Genève, Switzerland.,U1195, INSERM and University Paris Sud, 94276 Kremlin Bicêtre, France
| | - Philippe Liere
- U1195, INSERM and University Paris Sud, 94276 Kremlin Bicêtre, France
| | - Jean-Paul Oudinet
- U1195, INSERM and University Paris Sud, 94276 Kremlin Bicêtre, France
| | - Alexandre Hertig
- Department of Nephrology, Tenon Hospital, APHP, 75020 Paris, France.,University of Pierre and Marie Curie, Sorbonne University, Paris 06, 75005 Paris, France.,Unité Mixte de Recherche Scientifique 1155, F-75020 Paris, France
| | - Guillaume Lefèvre
- University of Pierre and Marie Curie, Sorbonne University, Paris 06, 75005 Paris, France.,Department of Biochemistry and Hormonology, Tenon Hospital, APHP, F-75020 Paris, France
| | - Nicola Pluchino
- Department of Gynecology and Obstetrics of University Hospital of Geneva, 1205, Genève, Switzerland
| | | | - Nathalie Chabbert-Buffet
- University of Pierre and Marie Curie, Sorbonne University, Paris 06, 75005 Paris, France.,Department of Obstetrics, Gynecology and Reproductive Medicine, Tenon Hospital, APHP, F-75020 Paris, France.,INSERM, UMR-S938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| |
Collapse
|
57
|
Trenti A, Tedesco S, Boscaro C, Ferri N, Cignarella A, Trevisi L, Bolego C. The Glycolytic Enzyme PFKFB3 Is Involved in Estrogen-Mediated Angiogenesis via GPER1. J Pharmacol Exp Ther 2017; 361:398-407. [PMID: 28348059 DOI: 10.1124/jpet.116.238212] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 03/22/2017] [Indexed: 01/08/2023] Open
Abstract
The endogenous estrogen 17β-estradiol (E2) is a key factor in promoting endothelial healing and angiogenesis. Recently, proangiogenic signals including vascular endothelial growth factor and others have been shown to converge in endothelial cell metabolism. Because inhibition of the glycolytic enzyme activator phosphofructokinase-2/fructose-2,6-bisphosphatase 3 (PFKFB3) reduces pathologic angiogenesis and estrogen receptor (ER) signaling stimulates glucose uptake and glycolysis by inducing PFKFB3 in breast cancer, we hypothesized that E2 triggers angiogenesis in endothelial cells via rapid ER signaling that requires PFKFB3 as a downstream effector. We report that treatment with the selective G protein-coupled estrogen receptor (GPER1) agonist G-1 (10-10 to 10-7 M) mimicked the chemotactic and proangiogenic effect of E2 as measured in a number of short-term angiogenesis assays in human umbilical vein endothelial cells (HUVECs); in addition, E2 treatment upregulated PFKFB3 expression in a time- and concentration-dependent manner. Such an effect peaked at 3 hours and was also induced by G-1 and abolished by pretreatment with the GPER1 antagonist G-15 or GPER1 siRNA, consistent with engagement of membrane ER. Experiments with the PFKFB3 inhibitor 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one showed that PFKFB3 activity was required for estrogen-mediated HUVEC migration via GPER1. In conclusion, E2-induced angiogenesis was mediated at least in part by the membrane GPER1 and required upregulation of the glycolytic activator PFKFB3 in HUVECs. These findings unravel a previously unrecognized mechanism of estrogen-dependent endocrine-metabolic crosstalk in HUVECs and may have implications in angiogenesis occurring in ischemic or hypoxic tissues.
Collapse
Affiliation(s)
- Annalisa Trenti
- Department of Pharmaceutical and Pharmacological Sciences (A.T., S.T., Ca.B., N.F., L.T., Ch.B) and Department of Medicine (A.C.), University of Padova, Padova, Italy
| | - Serena Tedesco
- Department of Pharmaceutical and Pharmacological Sciences (A.T., S.T., Ca.B., N.F., L.T., Ch.B) and Department of Medicine (A.C.), University of Padova, Padova, Italy
| | - Carlotta Boscaro
- Department of Pharmaceutical and Pharmacological Sciences (A.T., S.T., Ca.B., N.F., L.T., Ch.B) and Department of Medicine (A.C.), University of Padova, Padova, Italy
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences (A.T., S.T., Ca.B., N.F., L.T., Ch.B) and Department of Medicine (A.C.), University of Padova, Padova, Italy
| | - Andrea Cignarella
- Department of Pharmaceutical and Pharmacological Sciences (A.T., S.T., Ca.B., N.F., L.T., Ch.B) and Department of Medicine (A.C.), University of Padova, Padova, Italy
| | - Lucia Trevisi
- Department of Pharmaceutical and Pharmacological Sciences (A.T., S.T., Ca.B., N.F., L.T., Ch.B) and Department of Medicine (A.C.), University of Padova, Padova, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences (A.T., S.T., Ca.B., N.F., L.T., Ch.B) and Department of Medicine (A.C.), University of Padova, Padova, Italy
| |
Collapse
|
58
|
Regitz-Zagrosek V, Kararigas G. Mechanistic Pathways of Sex Differences in Cardiovascular Disease. Physiol Rev 2017; 97:1-37. [PMID: 27807199 DOI: 10.1152/physrev.00021.2015] [Citation(s) in RCA: 417] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Major differences between men and women exist in epidemiology, manifestation, pathophysiology, treatment, and outcome of cardiovascular diseases (CVD), such as coronary artery disease, pressure overload, hypertension, cardiomyopathy, and heart failure. Corresponding sex differences have been studied in a number of animal models, and mechanistic investigations have been undertaken to analyze the observed sex differences. We summarize the biological mechanisms of sex differences in CVD focusing on three main areas, i.e., genetic mechanisms, epigenetic mechanisms, as well as sex hormones and their receptors. We discuss relevant subtypes of sex hormone receptors, as well as genomic and nongenomic, activational and organizational effects of sex hormones. We describe the interaction of sex hormones with intracellular signaling relevant for cardiovascular cells and the cardiovascular system. Sex, sex hormones, and their receptors may affect a number of cellular processes by their synergistic action on multiple targets. We discuss in detail sex differences in organelle function and in biological processes. We conclude that there is a need for a more detailed understanding of sex differences and their underlying mechanisms, which holds the potential to design new drugs that target sex-specific cardiovascular mechanisms and affect phenotypes. The comparison of both sexes may lead to the identification of protective or maladaptive mechanisms in one sex that could serve as a novel therapeutic target in one sex or in both.
Collapse
Affiliation(s)
- Vera Regitz-Zagrosek
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Georgios Kararigas
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
59
|
Lu Q, Schnitzler GR, Vallaster CS, Ueda K, Erdkamp S, Briggs CE, Iyer LK, Jaffe IZ, Karas RH. Unliganded estrogen receptor alpha regulates vascular cell function and gene expression. Mol Cell Endocrinol 2017; 442:12-23. [PMID: 27888004 DOI: 10.1016/j.mce.2016.11.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/03/2016] [Accepted: 11/21/2016] [Indexed: 01/15/2023]
Abstract
The unliganded form of the estrogen receptor is generally thought to be inactive. Our prior studies, however, suggested that unliganded estrogen receptor alpha (ERα) exacerbates adverse vascular injury responses in mice. Here, we show that the presence of unliganded ERα decreases vascular endothelial cell (EC) migration and proliferation, increases smooth muscle cell (SMC) proliferation, and increases inflammatory responses in cultured ECs and SMCs. Unliganded ERα also regulates many genes in vascular ECs and mouse aorta. Activation of ERα by E2 reverses the cell physiological effects of unliganded ERα, and promotes gene regulatory effects that are predicted to counter the effects of unliganded ERα. These results reveal that the unliganded form of ERα is not inert, but significantly impacts gene expression and physiology of vascular cells. Furthermore, they indicate that the cardiovascular protective effects of estrogen may be connected to its ability to counteract these effects of unliganded ERα.
Collapse
Affiliation(s)
- Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Gavin R Schnitzler
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA.
| | - Caroline S Vallaster
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Kazutaka Ueda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Stephanie Erdkamp
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Christine E Briggs
- Tufts Center for Neuroscience Research, Neuroscience Department, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Lakshmanan K Iyer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Richard H Karas
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA.
| |
Collapse
|
60
|
Disrupting Tumor Angiogenesis and "the Hunger Games" for Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:171-195. [PMID: 29282684 DOI: 10.1007/978-981-10-6020-5_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Angiogenesis, one of the hallmarks of cancers, has become an attractive target for cancer therapy since decades ago. It is broadly thought that upregulation of angiogenesis is involved in tumor progression and metastasis. Though tumor vessels are tortuous, disorganized, and leaky, they deliver oxygen and nutrients for tumor development. Based on this knowledge, many kinds of drugs targeting angiogenesis pathways have been developed, such as bevacizumab. However, the clinical outcomes of anti-angiogenesis therapies are moderate in metastatic breast cancer as well as in metastatic colorectal cancer and non-small cell lung cancer, even combined with traditional chemotherapy. In this chapter, the morphologic angiogenesis patterns and the key molecular pathways regulating angiogenesis are elaborated. The FDA-approved anti-angiogenesis drugs and current challenges of anti-angiogenesis therapy are described. The strategies to overcome the barriers will also be elucidated.
Collapse
|
61
|
Novel Regulators of Hemodynamics in the Pregnant Uterus. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 145:181-216. [DOI: 10.1016/bs.pmbts.2016.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
62
|
Morgan MM, Johnson BP, Livingston MK, Schuler LA, Alarid ET, Sung KE, Beebe DJ. Personalized in vitro cancer models to predict therapeutic response: Challenges and a framework for improvement. Pharmacol Ther 2016; 165:79-92. [PMID: 27218886 PMCID: PMC5439438 DOI: 10.1016/j.pharmthera.2016.05.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Personalized cancer therapy focuses on characterizing the relevant phenotypes of the patient, as well as the patient's tumor, to predict the most effective cancer therapy. Historically, these methods have not proven predictive in regards to predicting therapeutic response. Emerging culture platforms are designed to better recapitulate the in vivo environment, thus, there is renewed interest in integrating patient samples into in vitro cancer models to assess therapeutic response. Successful examples of translating in vitro response to clinical relevance are limited due to issues with patient sample acquisition, variability and culture. We will review traditional and emerging in vitro models for personalized medicine, focusing on the technologies, microenvironmental components, and readouts utilized. We will then offer our perspective on how to apply a framework derived from toxicology and ecology towards designing improved personalized in vitro models of cancer. The framework serves as a tool for identifying optimal readouts and culture conditions, thus maximizing the information gained from each patient sample.
Collapse
Affiliation(s)
- Molly M Morgan
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Brian P Johnson
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Megan K Livingston
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Elaine T Alarid
- Department of Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Kyung E Sung
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States.
| | - David J Beebe
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States; Department of Oncology, University of Wisconsin-Madison, Madison, WI, United States.
| |
Collapse
|
63
|
Association of endothelial proliferation with the magnitude of weight loss during calorie restriction. Angiogenesis 2016; 19:407-19. [PMID: 27245991 DOI: 10.1007/s10456-016-9516-7] [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] [Received: 01/15/2016] [Accepted: 05/20/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Substantial weight loss through intense dietary regimens is thought to ameliorate endothelial dysfunction in obesity. It is less clear whether similar improvements can be achieved with modest dietary interventions. This study aimed to identify the parameters of endothelial cell status in obesity that are affected by mild calorie restriction. METHODS Human umbilical vein endothelial cells (EA.hy926 line) in culture were exposed pairwise to serum from 57 individuals with simple obesity (BMI > 30 kg/m(2)) collected before and after 8-week dietary intervention with energy deficit of 300-500 kcal/day. RESULTS Analysis of endothelial transcriptome suggested that the intervention could impact on endothelial cell growth. Cell proliferation was measured with the MTT test and verified by [(3)H]-thymidine incorporation. The participants were categorized according to a change in proliferation over time. Significant decrease in endothelial cell proliferation correlated with the extent of weight loss in men, but not in women. This effect corresponded with changes in serum levels of leptin and adiponectin, but was not related to serum concentrations of several known angiogenic mediators (VEGF, MCP-1, TSP-1, MMP-9, angiopoietin-2). CONCLUSION Direction and magnitude of changes in serum-induced endothelial cell proliferation identifies patients with the greatest weight loss in response to modest calorie restriction.
Collapse
|
64
|
Dickler MN, Barry WT, Cirrincione CT, Ellis MJ, Moynahan ME, Innocenti F, Hurria A, Rugo HS, Lake DE, Hahn O, Schneider BP, Tripathy D, Carey LA, Winer EP, Hudis CA. Phase III Trial Evaluating Letrozole As First-Line Endocrine Therapy With or Without Bevacizumab for the Treatment of Postmenopausal Women With Hormone Receptor-Positive Advanced-Stage Breast Cancer: CALGB 40503 (Alliance). J Clin Oncol 2016; 34:2602-9. [PMID: 27138575 DOI: 10.1200/jco.2015.66.1595] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To investigate whether anti-vascular endothelial growth factor therapy with bevacizumab prolongs progression-free survival (PFS) when added to first-line letrozole as treatment of hormone receptor-positive metastatic breast cancer (MBC). PATIENTS AND METHODS Women with hormone receptor-positive MBC were randomly assigned 1:1 in a multicenter, open-label, phase III trial of letrozole (2.5 mg orally per day) with or without bevacizumab (15 mg/kg intravenously once every 3 weeks) within strata defined by measurable disease and disease-free interval. This trial had 90% power to detect a 50% improvement in median PFS from 6 to 9 months. Using a one-sided α = .025, a target sample size of 352 patients was planned. RESULTS From May 2008 to November 2011, 350 women were recruited; 343 received treatment and were observed for efficacy and safety. Median age was 58 years (range, 25 to 87 years). Sixty-two percent had measurable disease, and 45% had de novo MBC. At a median follow-up of 39 months, the addition of bevacizumab resulted in a significant reduction in the hazard of progression (hazard ratio, 0.75; 95% CI, 0.59 to 0.96; P = .016) and a prolongation in median PFS from 15.6 months with letrozole to 20.2 months with letrozole plus bevacizumab. There was no significant difference in overall survival (hazard ratio, 0.87; 95% CI, 0.65 to 1.18; P = .188), with median overall survival of 43.9 months with letrozole versus 47.2 months with letrozole plus bevacizumab. The largest increases in incidence of grade 3 to 4 treatment-related toxicities with the addition of bevacizumab were hypertension (24% v 2%) and proteinuria (11% v 0%). CONCLUSION The addition of bevacizumab to letrozole improved PFS in hormone receptor-positive MBC, but this benefit was associated with a markedly increased risk of grade 3 to 4 toxicities. Research on predictive markers will be required to clarify the role of bevacizumab in this setting.
Collapse
Affiliation(s)
- Maura N Dickler
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN.
| | - William T Barry
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Constance T Cirrincione
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Matthew J Ellis
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Mary Ellen Moynahan
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Federico Innocenti
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Arti Hurria
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Hope S Rugo
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Diana E Lake
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Olwen Hahn
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Bryan P Schneider
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Debasish Tripathy
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Lisa A Carey
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Eric P Winer
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| | - Clifford A Hudis
- Maura N. Dickler, Mary Ellen Moynahan, Diana E. Lake, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center, New York, NY; William T. Barry, Dana-Farber Cancer Institute; Eric P. Winer, Dana-Farber/Partners Cancer Care, Boston, MA; Constance T. Cirrincione, Duke University, Durham, NC; Matthew J. Ellis, Baylor College of Medicine; Debasish Tripathy, The University of Texas MD Anderson Cancer Center, Houston, TX; Federico Innocenti and Lisa A. Carey, University of North Carolina at Chapel Hill, Chapel Hill, NC; Arti Hurria, City of Hope, Duarte; Hope S. Rugo, University of California at San Francisco, San Francisco, CA; Olwen Hahn, Alliance for Clinical Trials in Oncology, Chicago, IL; and Bryan P. Schneider, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
65
|
Shimizu T. Molecular and cellular mechanisms for the regulation of ovarian follicular function in cows. J Reprod Dev 2016; 62:323-9. [PMID: 27097851 PMCID: PMC5004786 DOI: 10.1262/jrd.2016-044] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ovary is an important organ that houses the oocytes
(reproductive cell). Oocyte growth depends on the
function of follicular cells such as the granulosa
and theca cells. Two-cell two gonadotropin systems
are associated with oocyte growth and follicular
cell functions. In addition to these systems, it
is also known that several growth factors regulate
oocyte growth and follicular cell functions.
Vascular endothelial growth factor (VEGF) is
involved in thecal vasculature during follicular
development and the suppression of granulosa cell
apoptosis. Metabolic factors such as insulin,
growth hormone (GH) and insulin-like growth factor
1 (IGF-1) also play critical roles in the process
of follicular development and growth. These
factors are associated not only with follicular
development, but also with follicular cell
function. Steroid hormones (estrogens, androgens,
and progestins) that are secreted from follicular
cells influence the function of the female genital
tract and its affect the susceptibility to
bacterial infection. This review covers our
current understanding of the mechanisms by which
gonadotrophins and/or steroid hormones regulate
the growth factors in the follicular cells of the
bovine ovary. In addition, this review describes
the effect of endotoxin on the function of
follicular cells.
Collapse
Affiliation(s)
- Takashi Shimizu
- Graduate School of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido 080-8555, Japan
| |
Collapse
|
66
|
Lu Q, Schnitzler GR, Ueda K, Iyer LK, Diomede OI, Andrade T, Karas RH. ER Alpha Rapid Signaling Is Required for Estrogen Induced Proliferation and Migration of Vascular Endothelial Cells. PLoS One 2016; 11:e0152807. [PMID: 27035664 PMCID: PMC4818104 DOI: 10.1371/journal.pone.0152807] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 02/25/2016] [Indexed: 01/08/2023] Open
Abstract
Estrogen promotes the proliferation and migration of vascular endothelial cells (ECs), which likely underlies its ability to accelerate re-endothelialization and reduce adverse remodeling after vascular injury. In previous studies, we have shown that the protective effects of E2 (the active endogenous form of estrogen) in vascular injury require the estrogen receptor alpha (ERα). ERα transduces the effects of estrogen via a classical DNA binding, "genomic" signaling pathway and via a more recently-described "rapid" signaling pathway that is mediated by a subset of ERα localized to the cell membrane. However, which of these pathways mediates the effects of estrogen on endothelial cells is poorly understood. Here we identify a triple point mutant version of ERα (KRR ERα) that is specifically defective in rapid signaling, but is competent to regulate transcription through the "genomic" pathway. We find that in ECs expressing wild type ERα, E2 regulates many genes involved in cell migration and proliferation, promotes EC migration and proliferation, and also blocks the adhesion of monocytes to ECs. ECs expressing KRR mutant ERα, however, lack all of these responses. These observations establish KRR ERα as a novel tool that could greatly facilitate future studies into the vascular and non-vascular functions of ERα rapid signaling. Further, they support that rapid signaling through ERα is essential for many of the transcriptional and physiological responses of ECs to E2, and that ERα rapid signaling in ECs, in vivo, may be critical for the vasculoprotective and anti-inflammatory effects of estrogen.
Collapse
Affiliation(s)
- Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Gavin R. Schnitzler
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- * E-mail: (GRS); (RHK)
| | - Kazutaka Ueda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Lakshmanan K. Iyer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Olga I. Diomede
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Tiffany Andrade
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Richard H. Karas
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- * E-mail: (GRS); (RHK)
| |
Collapse
|
67
|
Nikhil K, Sharan S, Wishard R, Palla SR, Krishna Peddinti R, Roy P. Pterostilbene carboxaldehyde thiosemicarbazone, a resveratrol derivative inhibits 17β-Estradiol induced cell migration and proliferation in HUVECs. Steroids 2016; 108:17-30. [PMID: 26850466 DOI: 10.1016/j.steroids.2016.01.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 01/09/2016] [Accepted: 01/29/2016] [Indexed: 11/23/2022]
Abstract
Angiogenesis plays important roles in tumor growth and metastasis, thus development of a novel angiogenesis inhibitor is essential for the improvement of therapeutics against cancer. Thrombospondins-1 (TSP-1) is a potent endogenous inhibitor of angiogenesis that acts through direct effects on endothelial cell migration, proliferation, survival, and activating apoptotic pathways. TSP-1 has been shown to disrupt estrogen-induced endothelial cell proliferation and migration. Here we investigated the potential of pterostilbene carboxaldehyde thiosemicarbazone (PTERC-T), a novel resveratrol (RESV) derivative, to inhibit angiogenesis induced by female sex steroids, particularly 17β-Estradiol (E2), on Human umbilical vein endothelial cells (HUVECs) and to elucidate the involvement of TSP-1 in PTERC-T action. Our results showed that PTERC-T significantly inhibited 17β-E2-stimulated proliferation of HUVECs and induced apoptosis as determined by annexin V/propidium iodide staining and cleaved caspase-3 expression. Furthermore, PTERC-T also inhibited endothelial cell migration, and invasion in chick chorioallantoic membrane (CAM) assay. In contrast, RESV failed to inhibit 17β-E2 induced HUVECs proliferation and invasion at similar dose. PTERC-T was also found to increase TSP-1 protein expression levels in a dose-dependent manner which, however, was counteracted by co-incubation with p38MAPK or JNK inhibitors, suggesting involvement of these pathways in PTERC-T action. These results suggest that the inhibitory effect of PTERC-T on 17β-E2 induced angiogenesis is associated, at least in part, with its induction of endothelial cell apoptosis and inhibition of cell migration through targeting TSP-1. Thus, PTERC-T could be considered as a potential lead compound for developing a class of new drugs targeting angiogenesis-related diseases.
Collapse
Affiliation(s)
- Kumar Nikhil
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | - Shruti Sharan
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | - Rohan Wishard
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | - Srinivasa Rao Palla
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | - Rama Krishna Peddinti
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| |
Collapse
|
68
|
Chakrabarti M, Das A, Samantaray S, Smith JA, Banik NL, Haque A, Ray SK. Molecular mechanisms of estrogen for neuroprotection in spinal cord injury and traumatic brain injury. Rev Neurosci 2016; 27:271-81. [DOI: 10.1515/revneuro-2015-0032] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 08/27/2015] [Indexed: 01/18/2023]
Abstract
AbstractEstrogen (EST) is a steroid hormone that exhibits several important physiological roles in the human body. During the last few decades, EST has been well recognized as an important neuroprotective agent in a variety of neurological disorders in the central nervous system (CNS), such as spinal cord injury (SCI), traumatic brain injury (TBI), Alzheimer’s disease, and multiple sclerosis. The exact molecular mechanisms of EST-mediated neuroprotection in the CNS remain unclear due to heterogeneity of cell populations that express EST receptors (ERs) in the CNS as well as in the innate and adaptive immune system. Recent investigations suggest that EST protects the CNS from injury by suppressing pro-inflammatory pathways, oxidative stress, and cell death, while promoting neurogenesis, angiogenesis, and neurotrophic support. In this review, we have described the currently known molecular mechanisms of EST-mediated neuroprotection and neuroregeneration in SCI and TBI. At the same time, we have emphasized on the recent in vitro and in vivo findings from our and other laboratories, implying potential clinical benefits of EST in the treatment of SCI and TBI.
Collapse
Affiliation(s)
- Mrinmay Chakrabarti
- 1Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29209, USA
| | - Arabinda Das
- 2Department of Neurosurgery and Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Supriti Samantaray
- 2Department of Neurosurgery and Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Joshua A. Smith
- 2Department of Neurosurgery and Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Naren L. Banik
- 2Department of Neurosurgery and Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Azizul Haque
- 3Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Swapan K. Ray
- 1Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29209, USA
| |
Collapse
|
69
|
Does hypoxia play a role in infantile hemangioma? Arch Dermatol Res 2016; 308:219-27. [PMID: 26940670 DOI: 10.1007/s00403-016-1635-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/13/2015] [Accepted: 02/15/2016] [Indexed: 12/17/2022]
Abstract
Infantile hemangioma (IH), the most common tumor of infancy, is characterized by rapid growth during infancy, followed by spontaneous involution over 5-10 years. Certain clinical observations have led to the suggestion that IH is triggered and maintained by hypoxia. We review the literature on the possible role of hypoxia in the etiology of IH, in particular, (1) the role of hypoxia inducible factor-1α (HIF-1α) and its downstream targets including GLUT-1 and VEGF; (2) the pathophysiological link between IH and retinopathy of prematurity; (3) hypoxic events in the early life including placental insufficiency, pre-eclampsia and low birthweight that have the potential to promote hypoxic stress; and (4) the evidence supporting the development of IH independent of HIF-1α. We also discuss these observations in the context of recent evidence of the crucial role of stem cells and the cytokines niche that governs their proliferation and inevitable differentiation, offering novel insights into the biology of IH. We propose that various triggers may simultaneously up-regulate HIF-1α, which is downstream of the renin-angiotensin system, specifically angiotensin II, which promotes production of HIF-1α. These developments shed light to the understanding of this enigmatic condition.
Collapse
|
70
|
Li S, Dang Y, Zhou X, Huang B, Huang X, Zhang Z, Kwan YW, Chan SW, Leung GPH, Lee SMY, Hoi MPM. Formononetin promotes angiogenesis through the estrogen receptor alpha-enhanced ROCK pathway. Sci Rep 2015; 5:16815. [PMID: 26568398 PMCID: PMC4645220 DOI: 10.1038/srep16815] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/14/2015] [Indexed: 11/09/2022] Open
Abstract
Formononetin is an isoflavone that has been shown to display estrogenic properties and induce angiogenesis activities. However, the interrelationship between the estrogenic properties and angiogenesis activities of formononetin are not well defined. In the present study, docking and enzymatic assay demonstrated that formononetin displayed direct binding to the ligand-binding domain (LBD) of estrogen receptor alpha (ERα) with an agonistic property. Results from Human Umbilical Vein Endothelial Cells (HUVEC) by using real-time migration xCELLigence system, immunofluorescence and western blotting provided strong evidences of formononetin induced endothelial cell migration and dramatic actin cytoskeleton spatial modification through ERα-enhanced-ROCK-II/MMP2/9 signaling pathways. In addition, results from co-immunoprecipitation suggested formononetin induced cell migration via recruiting of ERα/ROCK-II activated complex formation. More interestingly, in zebrafish embryo we observed that formononetin significantly promoted angiogenic sproutings in the subintestinal vessels (SIVs) that could be completely abolished by ROCK inhibitor. In this study, we elucidated the underlying mechanisms that formononetin produced proangiogenesis effects through an ERα-enhanced ROCK-II signaling pathways. Results from the present study also expand our knowledge about the enigmatic underlying mechanisms of phytoestrogenic compounds in the promotion of angiogenesis in relation to ERα and ROCK interaction in endothelial cells and their relationship with actin assembly and cell migration.
Collapse
Affiliation(s)
- Shang Li
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yuanye Dang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xuelin Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Bin Huang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xiaohui Huang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zherui Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yiu Wa Kwan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Shun Wan Chan
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - George Pak Heng Leung
- Pharmacology and Pharmacy, Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Simon Ming Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
71
|
Ambrose C. Muscle weakness during aging: a deficiency state involving declining angiogenesis. Ageing Res Rev 2015; 23:139-53. [PMID: 26093038 DOI: 10.1016/j.arr.2015.03.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 03/19/2015] [Accepted: 03/30/2015] [Indexed: 12/12/2022]
Abstract
This essay begins by proposing that muscle weakness of old age from sarcopenia is due in large part to reduced capillary density in the muscles, as documented in 9 reports of aged persons and animals. Capillary density (CD) is determined by local levels of various angiogenic factors, which also decline in muscles with aging, as reported in 7 studies of old persons and animals. There are also numerous reports of reduced CD in the aged brain and other studies showing reduced CD in the kidney and heart of aged animals. Thus a waning angiogenesis throughout the body may be a natural occurrence in later years and may account significantly for the lesser ailments (physical and cognitive) of elderly people. Old age is regarded here as a deficiency state which may be corrected by therapeutic angiogenesis, much as a hormonal deficiency can be relieved by the appropriate hormone therapy. Such therapy could employ recombinant angiogenic factors which are now commercially available.
Collapse
|
72
|
Pence JC, Clancy KBH, Harley BAC. The induction of pro-angiogenic processes within a collagen scaffold via exogenous estradiol and endometrial epithelial cells. Biotechnol Bioeng 2015; 112:2185-94. [PMID: 25944769 DOI: 10.1002/bit.25622] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 04/13/2015] [Indexed: 12/23/2022]
Abstract
Nutrient transport remains a major limitation in the design of biomaterials. One approach to overcome this constraint is to incorporate features to induce angiogenesis-mediated microvasculature formation. Angiogenesis requires a temporal presentation of both pro- and anti-angiogenic factors to achieve stable vasculature, leading to increasingly complex biomaterial design scheme. The endometrium, the lining of the uterus and site of embryo implantation, exemplifies a non-pathological model of rapid growth, shedding, and re-growth of dense vascular networks regulated by the dynamic actions of estradiol and progesterone. In this study, we examined the individual and combined response of endometrial epithelial cells and human umbilical vein endothelial cells to exogenous estradiol within a three-dimensional collagen scaffold. While endothelial cells did not respond to exogenous estradiol, estradiol directly stimulated endometrial epithelial cell transduction pathways and resulted in dose-dependent increases in endogenous VEGF production. Co-culture experiments using conditioned media demonstrated estradiol stimulation of endometrial epithelial cells can induce functional changes in endothelial cells within the collagen biomaterial. We also report the effect of direct endometrial epithelial and endothelial co-culture as well as covalent immobilization of estradiol within the collagen biomaterial. These efforts establish the suitability of an endometrial-inspired model for promoting pro-angiogenic events within regenerative medicine applications. These results also suggest the potential for developing biomaterial-based models of the endometrium.
Collapse
Affiliation(s)
- Jacquelyn C Pence
- Department of Chemical Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Kathryn B H Clancy
- Department of Anthropology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Brendan A C Harley
- Department of Chemical Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois. .,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801.
| |
Collapse
|
73
|
Shi W, Zhu LI, Wang Y, Hu B, Xiao H, Zhou G, Chen C. Estrogen therapy to treat retinopathy in newborn mice. Exp Ther Med 2015; 10:611-617. [PMID: 26622363 DOI: 10.3892/etm.2015.2554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/21/2015] [Indexed: 12/19/2022] Open
Abstract
The aim of the present study was to treat retinopathy of prematurity (ROP) with estrogen (E2) so as to elucidate the role of E2 in the pathogenesis of ROP. A total of 120 postnatal 7-day-old (P7) C57BL/6J mice were selected and raised in a high-oxygen environment (75% oxygen) for 5 days, followed by 5 days in normal room air. Different doses of E2 or normal saline (NS) were injected intraperitoneally during different time-periods, and the mice were divided into 14 groups according dose of E2 injection (0.5-1.5 µg/0.05 ml) and dosing time. Blood vessel changes and hyperplasia were evaluated in flat-mounted retina and retinal slices. All mice that were exposed to room air, whether they were administered E2 or NS, showed good vascular development in the flat-mounted retina at P17. No increase in the number of endothelial cell nuclei in the new blood vessels was observed. In ascending order of E2 dose the numbers of cell nuclei were as follows: 0.18±0.129, 0.28±0.086 and 0.55±0.110. The number in the NS group was 2.12±0.373. When the results of the room-air groups were compared with those of the hyperoxia groups, a highly significant difference was found in each comparison (P<0.0001). All mice showed varying degrees of neovascularization and vascular obstruction in the flat-mounted retina at P17, and it was difficult to compare the blood vessels morphologically among these groups. The number of endothelial cell nuclei decreased following E2 injection, and the difference from the NS group exposed to hyperoxia was highly significant (P<0.0001). For all dose levels, the number of cell nuclei was the lowest when the drug was administered during P7-16, and the difference from the other two time-periods was statistically significant (P<0.05). When E2 was administered during P7-16, the number of cell nuclei was 15.5±1.993 in the 0.5-µg group, 14.23±2.49 in the 1.0-µg group and 18.05±1.62 in the 1.5-µg group. No significant difference was found among these three groups (P>0.05). In conclusion, E2 treatment during the development of retinopathy can improve symptoms in neonatal mice, suggesting that E2 plays an important role at the two initial stages in the pathogenesis of ROP. This may indicate new pharmacological measures to prevent and treat ROP.
Collapse
Affiliation(s)
- Wenjing Shi
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - L I Zhu
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - Yuhuan Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - Baoyang Hu
- Department of Human Anatomy, Histology and Embryology, Fudan University, Shanghai 200032, P.R. China
| | - Honglei Xiao
- Department of Human Anatomy, Histology and Embryology, Fudan University, Shanghai 200032, P.R. China
| | - Guoming Zhou
- Department of Human Anatomy, Histology and Embryology, Fudan University, Shanghai 200032, P.R. China
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| |
Collapse
|
74
|
Electrospun polyurethane-dextran nanofiber mats loaded with Estradiol for post-menopausal wound dressing. Int J Biol Macromol 2015; 77:1-8. [DOI: 10.1016/j.ijbiomac.2015.02.044] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/15/2015] [Accepted: 02/15/2015] [Indexed: 12/13/2022]
|
75
|
Ishida N, Fukazawa T, Maeda Y, Yamatsuji T, Kato K, Matsumoto K, Shimo T, Takigawa N, Whitsett JA, Naomoto Y. A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor. Exp Cell Res 2015; 335:197-206. [PMID: 25839409 DOI: 10.1016/j.yexcr.2015.03.019] [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: 10/05/2014] [Revised: 03/04/2015] [Accepted: 03/23/2015] [Indexed: 12/31/2022]
Abstract
The PI3K-AKT pathway is expected to be a therapeutic target for non-small cell lung cancer (NSCLC) treatment. We previously reported that a novel PI3K inhibitor iMDK suppressed NSCLC cells in vitro and in vivo without harming normal cells and mice. Unexpectedly, iMDK activated the MAPK pathway, including ERK, in the NSCLC cells. Since iMDK did not eradicate such NSCLC cells completely, it is possible that the activated MAPK pathway confers resistance to the NSCLC cells against cell death induced by iMDK. In the present study, we assessed whether suppressing of iMDK-mediated activation of the MAPK pathway would enhance anti-tumorigenic activity of iMDK. PD0325901, a MAPK inhibitor, suppressed the MAPK pathway induced by iMDK and cooperatively inhibited cell viability and colony formation of NSCLC cells by inducing apoptosis in vitro. HUVEC tube formation, representing angiogenic processes in vitro, was also cooperatively inhibited by the combinatorial treatment of iMDK and PD0325901. The combinatorial treatment of iMDK with PD0325901 cooperatively suppressed tumor growth and tumor-associated angiogenesis in a lung cancer xenograft model in vivo. Here, we demonstrate a novel treatment strategy using iMDK and PD0325901 to eradicate NSCLC.
Collapse
Affiliation(s)
- Naomasa Ishida
- Department of General Surgery, Kawasaki Medical School, Okayama 700-8505, Japan
| | - Takuya Fukazawa
- Department of General Surgery, Kawasaki Medical School, Okayama 700-8505, Japan.
| | - Yutaka Maeda
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229-3039, United States
| | - Tomoki Yamatsuji
- Department of General Surgery, Kawasaki Medical School, Okayama 700-8505, Japan
| | - Katsuya Kato
- Department of Diagnostic Radiology 2, Kawasaki Medical School, Okayama 700-8505, Japan
| | - Kenichi Matsumoto
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Tsuyoshi Shimo
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Nagio Takigawa
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama 700-8505, Japan
| | - Jeffrey A Whitsett
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229-3039, United States
| | - Yoshio Naomoto
- Department of General Surgery, Kawasaki Medical School, Okayama 700-8505, Japan
| |
Collapse
|
76
|
Gibson DA, Greaves E, Critchley HOD, Saunders PTK. Estrogen-dependent regulation of human uterine natural killer cells promotes vascular remodelling via secretion of CCL2. Hum Reprod 2015; 30:1290-301. [PMID: 25820695 PMCID: PMC4498222 DOI: 10.1093/humrep/dev067] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 03/04/2015] [Indexed: 12/02/2022] Open
Abstract
STUDY QUESTION Does intrauterine biosynthesis of estrogen play an important role in early pregnancy by altering the function of uterine natural killer (uNK) cells? SUMMARY ANSWER Estrogens directly regulate the function of human uNK cells by increasing uNK cell migration and secretion of uNK cell-derived chemokine (C-C motif) ligand 2 (CCL2) that critically facilitates uNK-mediated angiogenesis. WHAT IS KNOWN ALREADY uNK cells are a phenotypically distinct population of tissue-resident immune cells that regulate vascular remodelling within the endometrium and decidua. Recently we discovered that decidualisation of human endometrial stromal cells results in the generation of an estrogen-rich microenvironment in areas of decidualised endometrium. We hypothesize that intrauterine biosynthesis of estrogens plays an important role in early pregnancy by altering the function of uNK cells. STUDY DESIGN, SIZE, DURATION This laboratory-based study used primary human uNK cells which were isolated from first trimester human decidua (n = 32). PARTICIPANTS/MATERIALS, SETTING, METHODS Primary uNK cells were isolated from first trimester human decidua using magnetic cell sorting. The impact of estrogens on uNK cell function was assessed. Isolated uNK cells were treated with estrone (E1, 10−8 M) or estradiol (E2, 10−8 M) alone or in combination with the anti-estrogen ICI 182 780 (ICI, 10−6 M). uNK cell motility was assessed by transwell migration assay and time-lapse microscopy. Expression of chemokine receptors was assessed by quantitative PCR (qPCR) and immunohistochemistry, and angiogenic factors were assessed by qPCR and cytokine array. Concentrations of CCL2 in supernatants were measured by enzyme-linked immunosorbent assay. Angiogenesis was assessed in a human endometrial endothelial cell network formation assay. MAIN RESULTS AND THE ROLE OF CHANCE Treatment with either E1 or E2 increased uNK cell migration (P = 0.0092 and P = 0.0063, respectively) compared with control. Co-administration of the anti-estrogen ICI blocked the effects of E1 and E2 on cell migration. Concentrations of C-X-C chemokine receptor type 4 (CXCR4) mRNA in uNK cells were increased by E2 treatment. The network formation assay revealed that conditioned media from uNK cells treated with E2 significantly increased human endometrial endothelial cell (HEEC) angiogenesis (P = 0.0029 versus control). Analysis of media from uNK cells treated with E2 using an antibody array identified CCL2 as the most abundant cytokine. Validation assays confirmed concentrations of CCL2 mRNA and protein were increased by E2 in uNK cells (P < 0.05 versus controls). Compared with the control, recombinant human CCL2 was found to increase HEEC network formation (P < 0.05) and neutralization of CCL2 in uNK conditioned media significantly decreased E2-dependent uNK-mediated network formation (P = 0.0006). LIMITATIONS, REASONS FOR CAUTION Our results are based on in vitro responses of primary human cells and we cannot be certain that similar mechanisms occur in vivo in humans. Primary human uNK cells were isolated from first trimester decidua at a range of gestations (8–12 weeks), which may be a source of variation. Primary human uNK cells from non-pregnant endometrium were not assessed and therefore the responses of uNK cells to E2 treatment described in this study may be distinct to uNK cells from first trimester decidua. WIDER IMPLICATIONS OF THE FINDINGS E2 is an essential regulator of reproductive competence. This study demonstrates a critical role for E2 in regulating cellular cross-talk within the endometrium during early pregnancy. We provide the first evidence that E2 directly regulates the function of human uNK cells by altering uNK cell migration and the secretion of uNK-derived angiogenic factors. We describe a novel mechanism of estrogen-dependent secretion of CCL2 which critically mediates uNK-dependent endometrial angiogenesis. Dysregulation of uNK cell function has been implicated in the aetiology of early implantation disorders and disorders of pregnancy. These novel findings provide unique insight into the regulation of uNK cell activity during the establishment of pregnancy in women and highlight key processes which may be targeted in future therapeutic strategies. STUDY FUNDING/COMPETING INTEREST(S) Studies undertaken in the authors' laboratory were supported by MRC Programme Grant G1100356/1 to P.T.K.S. The authors have no conflicts of interest to disclose.
Collapse
Affiliation(s)
- D A Gibson
- Medical Research Council Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - E Greaves
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - H O D Critchley
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - P T K Saunders
- Medical Research Council Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
77
|
Zhang HH, Lechuga TJ, Tith T, Wang W, Wing DA, Chen DB. S-nitrosylation of cofilin-1 mediates estradiol-17β-stimulated endothelial cytoskeleton remodeling. Mol Endocrinol 2015; 29:434-44. [PMID: 25635941 DOI: 10.1210/me.2014-1297] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Rapid nitric oxide (NO) production via endothelial NO synthase (eNOS) activation represents a major signaling pathway for the cardiovascular protective effects of estrogens; however, the pathways after NO biosynthesis that estrogens use to function remain largely unknown. Covalent adduction of a NO moiety to cysteines, termed S-nitrosylation (SNO), has emerged as a key route for NO to directly regulate protein function. Cofilin-1 (CFL1) is a small actin-binding protein essential for actin dynamics and cytoskeleton remodeling. Despite being identified as a major SNO protein in endothelial cells, whether SNO regulates CFL-1 function is unknown. We hypothesized that estradiol-17β (E2β) stimulates SNO of CFL1 via eNOS-derived NO and that E2β-induced SNO-CFL1 mediates cytoskeleton remodeling in endothelial cells. Point mutation studies determined Cys80 as the primary SNO site among the 4 cysteines (Cys39/80/139/147) in CFL1. Substitutions of Cys80 with Ala or Ser were used to prepare the SNO-mimetic/deficient (C80A/S) CFL1 mutants. Recombinant wild-type (wt) and mutant CFL1 proteins were prepared; their actin-severing activity was determined by real-time fluorescence imaging analysis. The activity of C80A CFL1 was enhanced to that of the constitutively active S3/A CFL1, whereas the other mutants had no effects. C80A/S mutations lowered Ser3 phosphorylation. Treatment with E2β increased filamentous (F)-actin and filopodium formation in endothelial cells, which were significantly reduced in cells overexpressing wt-CFL. Overexpression of C80A, but not C80S, CFL1 decreased basal F-actin and further suppressed E2β-induced F-actin and filopodium formation compared with wt-CFL1 overexpression. Thus, SNO(Cys80) of cofilin-1 via eNOS-derived NO provides a novel pathway for mediating estrogen-induced endothelial cell cytoskeleton remodeling.
Collapse
Affiliation(s)
- Hong-hai Zhang
- Departments of Obstetrics and Gynecology (H-h.Z., T.J.L., T.T., W.W., D.A.W., D-b.C.) and Pathology (T.J.L., D-b.C.), University of California, Irvine, Irvine, California 92697
| | | | | | | | | | | |
Collapse
|
78
|
Mayra PR, Rosalina VL, López G, Iruretagoyena J, Magness R. [Regulation of uterine blood flow. II. Functions of estrogen and estrogen receptor α/β in genomic and non-genomic actions of the uterine endothelium]. ACTA ACUST UNITED AC 2014; 79:218-228. [PMID: 26113751 DOI: 10.4067/s0717-75262014000300011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Pregnancy is marked by changes and cardiovascular adaptations that are important for the maintenance and growth of the placenta and fetus. During this period, the uterine vascular adaptations manifest changes that can be classified as short or long term and they related to adaptations for vasodilation, angiogenic or remodeling. Estrogen and the classical estrogen receptors (ERs), ER-α and ER-β, have been shown to be partially responsible for facilitating this dramatic increase in uterine blood flow needed during pregnancy. This literature review discusses the basis for structural diversity and functional selectivity of ERs by estrogen, the role of ERs on the genomic and non-genomic effects in endothelial cells of uterine arteries (UAEC). These themes integrate scientific knowledge about the molecular regulation of UAEC to maintain the physiological increase in uteroplacental perfusion observed during normal pregnancy.
Collapse
Affiliation(s)
- Pastore R Mayra
- Programa de Endocrinología y Fisiología Reproductiva, Universidad de Wisconsin-Madison, Wisconsin, USA ; Laboratorio de Investigación Perinatal, Departamento de Obstetricia y Ginecología y División de Ciencias de la Reproducción, Universidad de Wisconsin-Madison, Wisconsin, USA
| | - Villalón L Rosalina
- Programa de Endocrinología y Fisiología Reproductiva, Universidad de Wisconsin-Madison, Wisconsin, USA ; Laboratorio de Investigación Perinatal, Departamento de Obstetricia y Ginecología y División de Ciencias de la Reproducción, Universidad de Wisconsin-Madison, Wisconsin, USA
| | - Gladys López
- Laboratorio de Investigación Perinatal, Departamento de Obstetricia y Ginecología y División de Ciencias de la Reproducción, Universidad de Wisconsin-Madison, Wisconsin, USA ; Departamento Perinatal, Universidad de Wisconsin-Madison, Wisconsin, USA
| | - Jesús Iruretagoyena
- Departamento Perinatal, Universidad de Wisconsin-Madison, Wisconsin, USA ; Departamento de Obstetricia y Ginecología de la División de Medicina Materno-Fetal. Universidad de Wisconsin-Madison, Wisconsin, USA
| | - Ronald Magness
- Programa de Endocrinología y Fisiología Reproductiva, Universidad de Wisconsin-Madison, Wisconsin, USA ; Laboratorio de Investigación Perinatal, Departamento de Obstetricia y Ginecología y División de Ciencias de la Reproducción, Universidad de Wisconsin-Madison, Wisconsin, USA ; Departamento Perinatal, Universidad de Wisconsin-Madison, Wisconsin, USA ; Departamento de Ciencias de Animales, Universidad de Wisconsin-Madison, Wisconsin, USA
| |
Collapse
|
79
|
Rooke TW, Felty CL. A different way to look at varicose veins. J Vasc Surg Venous Lymphat Disord 2014; 2:207-11. [PMID: 26993192 DOI: 10.1016/j.jvsv.2013.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/20/2013] [Accepted: 08/21/2013] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The development of varicose veins is commonly attributed to vessel wall degeneration. The idea that varicose veins occur because of pathological processes, however, is challenged by certain observations. For example, their high prevalence (50% or greater) in many populations makes it statistically "normal" to have varicose veins; their well-established genetic predisposition raises the possibility that this high prevalence reflects a survival benefit. One way to explain this apparent contradiction is to theorize that varicose veins are produced by the same mechanism(s) that lead to the growth and remodeling of other types of blood vessels. If so, being "good" at forming varicose veins may also predispose to being "good" at forming various types of collateral blood vessels when necessary. METHODS A selected literature review was conducted. Works chosen for review included those suggesting that: the process of varicose vein formation may share the same basic mechanisms as the formation of collateral veins, arteries, and lymphatic vessels; and clinical outcomes may be different between subjects with and without varicose veins. RESULTS Evidence suggests that subjects who are "good" at forming varicose veins may also be "good" at forming various types of collateral vessels, and they may have better overall survival (with less cardiovascular morbidity) than those without varicose veins. CONCLUSIONS Varicose veins may be "the price we pay" for an enhanced ability to form collateral vessels when necessary.
Collapse
Affiliation(s)
- Thom W Rooke
- Department of Vascular Medicine, Mayo Clinic, Rochester, Minn.
| | - Cindy L Felty
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minn
| |
Collapse
|
80
|
Yurdagul A, Kleinedler JJ, McInnis MC, Khandelwal AR, Spence AL, Orr AW, Dugas TR. Resveratrol promotes endothelial cell wound healing under laminar shear stress through an estrogen receptor-α-dependent pathway. Am J Physiol Heart Circ Physiol 2014; 306:H797-806. [PMID: 24464753 DOI: 10.1152/ajpheart.00892.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Restenosis is an adverse outcome of angioplasty, characterized by vascular smooth muscle cell (VSMC) hyperplasia. However, therapies targeting VSMC proliferation delay re-endothelialization, increasing the risk of thrombosis. Resveratrol (RESV) inhibits restenosis and promotes re-endothelialization after arterial injury, but in vitro studies assessing RESV-mediated effects on endothelial cell growth contradict these findings. We thus hypothesized that fluid shear stress, mimicking physiological blood flow, would recapitulate RESV-dependent endothelial cell wound healing. Since RESV is an estrogen receptor (ER) agonist, we tested whether RESV promotes re-endothelialization through an ER-α-dependent mechanism. Mice fed a high-fat diet or a diet supplemented with RESV were subjected to carotid artery injury. At 7 days after injury, RESV significantly accelerated re-endothelialization compared with vehicle. In vitro wound healing assays demonstrated that RESV exhibits cell-type selectivity, inhibiting VSMC, but not endothelial cell growth. Under laminar shear stress (LSS), RESV dramatically enhanced endothelial cell wound healing and increased both the activation of extracellular signal-regulated kinase (ERK) and endothelial cell proliferation. Under LSS, small interfering RNA against ER-α, but not endothelial nitric oxide synthase, abolished RESV-induced ERK activation, endothelial cell proliferation, and wound healing. Thus these studies suggest that the EC phenotype induced by LSS better models the prohealing effects of RESV and that RESV and LSS interact to promote an ER-α-dependent mitogenic effect in endothelial cells.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | | | | | | | | | | | | |
Collapse
|
81
|
Mahmoodzadeh S, Leber J, Zhang X, Jaisser F, Messaoudi S, Morano I, Furth PA, Dworatzek E, Regitz-Zagrosek V. Cardiomyocyte-specific Estrogen Receptor Alpha Increases Angiogenesis, Lymphangiogenesis and Reduces Fibrosis in the Female Mouse Heart Post-Myocardial Infarction. ACTA ACUST UNITED AC 2014; 5:153. [PMID: 24977106 DOI: 10.4172/2157-7013.1000153] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Experimental studies showed that 17β-estradiol (E2) and activated Estrogen Receptors (ER) protect the heart from ischemic injury. However, the underlying molecular mechanisms are not well understood. To investigate the role of ER-alpha (ERα) in cardiomyocytes in the setting of myocardial ischemia, we generated transgenic mice with cardiomyocyte-specific overexpression of ERα (ERα-OE) and subjected them to Myocardial Infarction (MI). At the basal level, female and male ERα-OE mice showed increased Left Ventricular (LV) mass, LV volume and cardiomyocyte length. Two weeks after MI, LV volume was significantly increased and LV wall thickness decreased in female and male WT-mice and male ERα-OE, but not in female ERα-OE mice. ERα-OE enhanced expression of angiogenesis and lymphangiogenesis markers (Vegf, Lyve-1), and neovascularization in the peri-infarct area in both sexes. However, attenuated level of fibrosis and higher phosphorylation of JNK signaling pathway could be detected only in female ERα-OE after MI. In conclusion, our study indicates that ERα protects female mouse cardiomyocytes from the sequelae of ischemia through induction of neovascularization in a paracrine fashion and impaired fibrosis, which together may contribute to the attenuation of cardiac remodelling.
Collapse
Affiliation(s)
- Shokoufeh Mahmoodzadeh
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin, Berlin, Germany
| | - Joachim Leber
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin, Berlin, Germany
| | - Xiang Zhang
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin, Berlin, Germany.,Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | | | - Ingo Morano
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Priscilla A Furth
- Departments of Oncology and Medicine and the Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Elke Dworatzek
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin, Berlin, Germany
| | - Vera Regitz-Zagrosek
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin, Berlin, Germany
| |
Collapse
|
82
|
Strategies to overcome endocrine therapy resistance in hormone receptor-positive advanced breast cancer. ACTA ACUST UNITED AC 2014. [DOI: 10.4155/cli.13.123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
83
|
Zheng Y, Feng Z, You C, Jin Y, Hu X, Wang X, Han C. In vitro evaluation of Panax notoginseng Rg1 released from collagen/chitosan-gelatin microsphere scaffolds for angiogenesis. Biomed Eng Online 2013; 12:134. [PMID: 24380420 PMCID: PMC3937171 DOI: 10.1186/1475-925x-12-134] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 12/26/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The emergence of skin substitutes provides a new approach for the treatment of wound repair and healing. The consistent and steady release of angiogenic factors is an important factor in the promotion of angiogenesis in skin substitutes, which usually lack, yet need, a vascular network. METHODS In this study, ginsenoside Rg1, a natural compound isolated from Panax notoginseng (PNS), was incorporated into a collagen/chitosan-gelatin microsphere (CC-GMS) scaffold. The cumulative release kinetics were evaluated, and the effects of the released Rg1 on human umbilical vein endothelial cells (HUVECs) behavior, including proliferation, migration, tube formation, cell-cycle progression, cell apoptosis, and vascular endothelial growth factor (VEGF) secretion, were investigated. Additionally, HUVECs were cultured on the CC-GMS scaffold to test its biocompatibility. Standard Rg1 and VEGF were used as positive controls. RESULTS The results indicated that the CC-GMS scaffold had good release kinetics. The Rg1 released from the CC-GMS scaffold did not lose its activity and had a significant effect on HUVEC proliferation. Both Rg1 and VEGF promoted HUVEC migration and tube formation. Rg1 did not induce HUVEC apoptosis but instead promoted HUVEC progression into the S and G2/M phases of the cell cycle. Rg1 significantly increased VEGF secretion compared with that in the control group. HUVEC culture on the CC-GMS scaffold indicated that this scaffold has good biocompatibility and that CC-GMS scaffolds containing different concentrations of Rg1 promote HUVEC attachment in a dose- and time-dependent manner. CONCLUSIONS Rg1 may represent a new class of angiogenic agent that can be encapsulated in CC-GMS scaffolds to exert angiogenic effects in engineered tissue.
Collapse
Affiliation(s)
- Yurong Zheng
- Department of Burns, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
| | - Zhanzeng Feng
- Department of Burns, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
| | - Chuangang You
- Department of Burns, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
| | - Yunyun Jin
- Department of Burns, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
| | - Xinlei Hu
- Department of Orthopedics, Second Affiliated Hospital of Zhejiang University, College of Medicine, Binjiang Branch, Hangzhou 310000, China
| | - Xingang Wang
- Department of Burns, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
| | - Chunmao Han
- Department of Burns, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
| |
Collapse
|
84
|
Markiewicz M, Richard E, Marks N, Ludwicka-Bradley A. Impact of endothelial microparticles on coagulation, inflammation, and angiogenesis in age-related vascular diseases. J Aging Res 2013; 2013:734509. [PMID: 24288612 PMCID: PMC3830876 DOI: 10.1155/2013/734509] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 09/04/2013] [Indexed: 11/17/2022] Open
Abstract
Endothelial microparticles (EMPs) are complex vesicular structures that originate from plasma membranes of activated or apoptotic endothelial cells. EMPs play a significant role in vascular function by altering the processes of inflammation, coagulation, and angiogenesis, and they are key players in the pathogenesis of several vascular diseases. Circulating EMPs are increased in many age-related vascular diseases such as coronary artery disease, peripheral vascular disease, cerebral ischemia, and congestive heart failure. Their elevation in plasma has been considered as both a biomarker and bioactive effector of vascular damage and a target for vascular diseases. This review focuses on the pleiotropic roles of EMPs and the mechanisms that trigger their formation, particularly the involvement of decreased estrogen levels, thrombin, and PAI-1 as major factors that induce EMPs in age-related vascular diseases.
Collapse
Affiliation(s)
- Margaret Markiewicz
- Division of Rheumatology and Immunology, Medical University of South Carolina, 114 Doughty Street, STB, Charleston, SC 29425, USA
| | - Erin Richard
- Department of Biology, College of Charleston, Rita Liddy Hollings Science Center, Charleston, SC 29424, USA
| | - Natalia Marks
- Department of Radiology, Maimonides Medical Center, Brooklyn, NY 11219, USA
| | - Anna Ludwicka-Bradley
- Division of Rheumatology and Immunology, Medical University of South Carolina, 114 Doughty Street, STB, Charleston, SC 29425, USA
| |
Collapse
|
85
|
Caliceti C, Aquila G, Pannella M, Morelli MB, Fortini C, Pinton P, Bonora M, Hrelia S, Pannuti A, Miele L, Rizzo P, Ferrari R. 17β-estradiol enhances signalling mediated by VEGF-A-delta-like ligand 4-notch1 axis in human endothelial cells. PLoS One 2013; 8:e71440. [PMID: 23967210 PMCID: PMC3742772 DOI: 10.1371/journal.pone.0071440] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 06/30/2013] [Indexed: 01/22/2023] Open
Abstract
Estrogens play a protective role in coronary artery disease. The mechanisms of action are still poorly understood, although a role for estrogens in stimulation of angiogenesis has been suggested. In several cell types, estrogens modulate the Notch pathway, which is involved in controlling angiogenesis downstream of vascular endothelial growth factor A (VEGF-A). The goal of our study was to establish whether estrogens modulate Notch activity in endothelial cells and the possible consequences on angiogenesis. Human umbilical vein endothelial cells (HUVECs) were treated with 17β-estradiol (E2) and the effects on Notch signalling were evaluated. E2 increased Notch1 processing as indicated by i) decreased levels of Notch1 transmembrane subunit ii) increased amount of Notch1 in nuclei iii) unaffected level of mRNA. Similarly, E2 increased the levels of the active form of Notch4 without altering Notch4 mRNA. Conversely, protein and mRNA levels of Notch2 were both reduced suggesting transcriptional repression of Notch2 by E2. Under conditions where Notch was activated by upregulation of Delta-like ligand 4 (Dll4) following VEGF-A treatment, E2 caused a further increase of the active form of Notch1, of the number of cells with nuclear Notch1 and of Hey2 mRNA. Estrogen receptor antagonist ICI 182.780 antagonized these effects suggesting that E2 modulation of Notch1 is mediated by estrogen receptors. E2 treatment abolished the increase in endothelial cells sprouting caused by Notch inhibition in a tube formation assay on 3D Matrigel and in mouse aortic ring explants. In conclusion, E2 affects several Notch pathway components in HUVECs, leading to an activation of the VEGF-A-Dll4-Notch1 axis and to a modulation of vascular branching when Notch signalling is inhibited. These results contribute to our understanding of the molecular mechanisms of cardiovascular protection exerted by estrogens by uncovering a novel role of E2 in the Notch signalling-mediated modulation of angiogenesis.
Collapse
Affiliation(s)
- Cristiana Caliceti
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Giorgio Aquila
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Micaela Pannella
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Marco Bruno Morelli
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Cinzia Fortini
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Surgeon, Morphology and Experimental Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Massimo Bonora
- Department of Surgeon, Morphology and Experimental Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Antonio Pannuti
- University of Mississippi, Medical Center Cancer Institute, Jackson, Mississippi, United States of America
| | - Lucio Miele
- University of Mississippi, Medical Center Cancer Institute, Jackson, Mississippi, United States of America
| | - Paola Rizzo
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| |
Collapse
|
86
|
Wen J, Zhao Y, Li J, Weng C, Cai J, Yang K, Yuan H, Imperato-McGinley J, Zhu YS. Suppression of DHT-induced paracrine stimulation of endothelial cell growth by estrogens via prostate cancer cells. Prostate 2013; 73:1069-81. [PMID: 23423946 PMCID: PMC3923318 DOI: 10.1002/pros.22654] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 01/23/2013] [Indexed: 11/08/2022]
Abstract
BACKGROUND Androgen modulation of angiogenesis in prostate cancer may be not directly mediated by androgen receptor (AR) as AR is not detected in the prostatic endothelial cells. METHODS We examined the paracrine stimulation of cell proliferation by prostate tumor cells and its modulation by androgen and estrogens in a murine endothelial cell line (MEC) that does not express AR. RESULTS Tumor cell conditioned media (TCM) collected from LAPC-4 or LNCaP prostatic tumor cells produced a time- and concentration-dependent induction of cell growth in MECs, which was parallel to the VEGF concentration in the TCM. This TCM-induced cell growth in MECs was enhanced by the treatment of prostatic tumor cells with dihydrotestosterone (DHT). Both the TCM-stimulation and DHT-enhancement effects in MECs were completely blocked by SU5416, a specific VEGF receptor antagonist. Co-administration of 17α-estradiol or 17β-estradiol with DHT in prostatic tumor cells completely inhibited the DHT-enhancement effect while treatment with DHT, 17α-estradiol or 17β-estradiol did not produce any significant direct effect in MECs. Moreover, administration of 17α-estradiol or 17β-estradiol in xenograft animals with LAPC-4 or LNCaP prostate tumor significantly decreased the microvessel number in the tumor tissues. CONCLUSIONS Our study indicated that prostate tumor cells regulate endothelial cell growth through a paracrine mechanism, which is mainly mediated by VEGF; and DHT is able to modulate endothelial cell growth via tumor cells, which is inhibited by 17α-estradiol and 17β-estradiol. Thus, both17α-estradiol and 17β-estradiol are potential agents for anti-angiogenesis therapy in androgen-responsive prostate cancer.
Collapse
Affiliation(s)
- Juan Wen
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
- The Center of Clinical Pharmacology, Central South
University, Changsha, China
| | - Yuan Zhao
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
| | - Jinghe Li
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
| | - Chunyan Weng
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
- The Center of Clinical Pharmacology, Central South
University, Changsha, China
| | - Jingjing Cai
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
- The Center of Clinical Pharmacology, Central South
University, Changsha, China
| | - Kan Yang
- Department of Cardiology of the Third Xiangya Hospital,
Central South University, Changsha, China
| | - Hong Yuan
- The Center of Clinical Pharmacology, Central South
University, Changsha, China
| | | | - Yuan-Shan Zhu
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
- The Institute of Clinical Pharmacology, Central South
University, Changsha, China
- Correspondence to: Yuan-Shan Zhu, Department of Medicine, Weill
Cornell Medical College, 1300 York Avenue, Box 149, New York, NY 10065.
| |
Collapse
|
87
|
Raeside JI, Christie HL, Waelchli RO, Betteridge KJ. Biosynthesis of oestrogen by the early equine embryo proper. Reprod Fertil Dev 2013; 24:1071-8. [PMID: 22950882 DOI: 10.1071/rd11275] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 03/07/2012] [Indexed: 11/23/2022] Open
Abstract
The embryo proper in early equine pregnancy has recently been shown to have a remarkable capacity for metabolism of oestrogens. High concentrations of oestrogens in yolk-sac fluid could provide substrate for local metabolism in tissues of the embryo proper and this activity could have significance for early development. Due to the high level of oestrogen metabolism in the embryo proper we examined the possibility that it could also biosynthesise oestrogens. Conceptuses were collected in the fourth week of pregnancy (n=23) and the embryo was separated from extraembryonic tissues for incubation with [(3)H]androstenedione. Steroids were recovered from media by solid-phase extraction and eluted as unconjugated and conjugated fractions. Profiles of free and sulfoconjugated fractions, as well as the phenolic steroids extracted from them, were obtained by chromatography. Oestrone and oestradiol were seen clearly, indicating oestrogen biosynthesis, and the presence of more polar products, arising from metabolism of the primary oestrogens, gave further evidence that the embryo was capable of oestrogen biosynthesis. Aromatase activity was also demonstrated by detection of tritium loss, as (3)H(2)O, from incubations (n=3) with [1β-(3)H]androstenedione. It is suggested that its oestrogen biosynthesis may have significance for the remarkable development of the vasculature in the embryo proper at this stage.
Collapse
Affiliation(s)
- James I Raeside
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G2W1, Canada.
| | | | | | | |
Collapse
|
88
|
Verbridge SS, Chakrabarti A, DelNero P, Kwee B, Varner JD, Stroock AD, Fischbach C. Physicochemical regulation of endothelial sprouting in a 3D microfluidic angiogenesis model. J Biomed Mater Res A 2013; 101:2948-56. [PMID: 23559519 DOI: 10.1002/jbm.a.34587] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 12/13/2012] [Accepted: 12/17/2012] [Indexed: 01/07/2023]
Abstract
Both physiological and pathological tissue remodeling (e.g., during wound healing and cancer, respectively) require new blood vessel formation via angiogenesis, but the underlying microenvironmental mechanisms remain poorly defined due in part to the lack of biologically relevant in vitro models. Here, we present a biomaterials-based microfluidic 3D platform for analysis of endothelial sprouting in response to morphogen gradients. This system consists of three lithographically defined channels embedded in type I collagen hydrogels. A central channel is coated with endothelial cells, and two parallel side channels serve as a source and a sink for the steady-state generation of biochemical gradients. Gradients of vascular endothelial growth factor (VEGF) promoted sprouting, whereby endothelial cell responsiveness was markedly dependent on cell density and vessel geometry regardless of treatment conditions. These results point toward mechanical and/or autocrine mechanisms that may overwhelm pro-angiogenic paracrine signaling under certain conditions. To date, neither geometrical effects nor cell density have been considered critical determinants of angiogenesis in health and disease. This biomimetic vessel platform demonstrated utility for delineating hitherto underappreciated contributors of angiogenesis, and future studies may enable important new mechanistic insights that will inform anti-angiogenic cancer therapy.
Collapse
Affiliation(s)
- Scott S Verbridge
- Department of Biomedical Engineering, Cornell University, Ithaca, New York
| | | | | | | | | | | | | |
Collapse
|
89
|
Liu S, Kilic G, Meyers MS, Navarro G, Wang Y, Oberholzer J, Mauvais-Jarvis F. Oestrogens improve human pancreatic islet transplantation in a mouse model of insulin deficient diabetes. Diabetologia 2013; 56:370-81. [PMID: 23132340 PMCID: PMC3536964 DOI: 10.1007/s00125-012-2764-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 10/12/2012] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS Pancreatic islet transplantation (PIT) offers a physiological treatment for type 1 diabetes, but the failure of islet engraftment hinders its application. The female hormone 17β-oestradiol (E2) favours islet survival and stimulates angiogenesis, raising the possibility that E2 may enhance islet engraftment following PIT. METHODS To explore this hypothesis, we used an insulin-deficient model with xenotransplantation of a marginal dose of human islets in nude mice rendered diabetic with streptozotocin. This was followed by 4 weeks of treatment with vehicle, E2, the non-feminising oestrogen 17α-oestradiol (17α-E2), the oestrogen receptor (ER) α agonist propyl-pyrazole-triol (PPT), the ERβ agonist diarylpropionitrile (DPN) or the G protein-coupled oestrogen receptor (GPER) agonist G1. RESULTS Treatment with E2, 17α-E2, PPT, DPN or G1 acutely improved blood glucose and eventually promoted islet engraftment, thus reversing diabetes. The effects of E2 were retained in the presence of immunosuppression and persisted after discontinuation of E2 treatment. E2 produced an acute decrease in graft hypoxic damage and suppressed beta cell apoptosis. E2 also acutely suppressed hyperglucagonaemia without altering insulin secretion, leading to normalisation of blood glucose. CONCLUSIONS/INTERPRETATION During PIT, E2 synergistic actions contribute to enhancing human islet-graft survival, revascularisation and functional mass. This study identifies E2 as a short-term treatment to improve PIT.
Collapse
Affiliation(s)
- S. Liu
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Tarry 15-761, Chicago, IL 60611 USA
- Present Address: Diabetes Institute, the First Affiliated Hospital of Xiamen University, Xiamen, China
| | - G. Kilic
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Tarry 15-761, Chicago, IL 60611 USA
| | - M. S. Meyers
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Tarry 15-761, Chicago, IL 60611 USA
| | - G. Navarro
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Tarry 15-761, Chicago, IL 60611 USA
| | - Y. Wang
- Department of Surgery, Division of Transplant Surgery, University of Illinois at Chicago, Chicago, IL USA
| | - J. Oberholzer
- Department of Surgery, Division of Transplant Surgery, University of Illinois at Chicago, Chicago, IL USA
| | - F. Mauvais-Jarvis
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Tarry 15-761, Chicago, IL 60611 USA
- Northwestern Comprehensive Center on Obesity, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| |
Collapse
|
90
|
Zhao J, Imbrie GA, Baur WE, Iyer LK, Aronovitz MJ, Kershaw TB, Haselmann GM, Lu Q, Karas RH. Estrogen receptor-mediated regulation of microRNA inhibits proliferation of vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2012; 33:257-65. [PMID: 23175673 DOI: 10.1161/atvbaha.112.300200] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Estradiol (E2) regulates gene transcription by activating estrogen receptor-α and estrogen receptor-β. Many of the genes regulated by E2 via estrogen receptors are repressed, yet the molecular mechanisms that mediate E2-induced gene repression are currently unknown. We hypothesized that E2, acting through estrogen receptors, regulates expression of microRNAs (miRs) leading to repression of expression of specific target genes. METHODS AND RESULTS Here, we report that E2 significantly upregulates the expression of 26 miRs and downregulates the expression of 6 miRs in mouse aorta. E2-mediated upregulation of one of these miRs, miR-203, was chosen for further study. In cultured vascular smooth muscle cells (VSMC), E2-mediated upregulation of miR-203 is mediated by estrogen receptor-α (but not estrogen receptor-β) via transcriptional upregulation of the primary miR. We demonstrate that the transcription factors Zeb-1 and AP-1 play critical roles in mediating E2-induced upregulation of miR-203 transcription. We show further that miR-203 mediates E2-induced repression of Abl1, and p63 protein abundance in VSMC. Finally, knocking-down miR-203 abolishes E2-mediated inhibition of VSMC proliferation, and overexpression of miR-203 inhibits cultured VSMC proliferation, but not vascular endothelial cell proliferation. CONCLUSIONS Our findings demonstrate that E2 regulates expression of miRs in the vasculature and support the estrogen receptors-dependent induction of miRs as a mechanism for E2-mediated gene repression. Furthermore, our findings demonstrate that miR-203 contributes to E2-induced inhibition of VSMC proliferation and highlight the potential of miR-203 as a therapeutic agent in the treatment of proliferative cardiovascular diseases.
Collapse
Affiliation(s)
- Jin Zhao
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Lohmann AE, Chia S. Patients with metastatic breast cancer using bevacizumab as a treatment: is there still a role for it? Curr Treat Options Oncol 2012; 13:249-62. [PMID: 22350496 DOI: 10.1007/s11864-012-0181-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OPINION STATEMENT Over the last few decades, the angiogenesis mechanism has increasingly been studied and implicated in cancer pathophysiology. At present, it is known that angiogenesis plays a relevant role in tumor growth, and more importantly many new molecules exists can potentially interfere with this process. Bevacizumab, a humanized monoclonal antibody targeting the vascular endothelial growth factor A (VEGF-A) now commonly used in the treatment of colorectal, renal cell, and brain cancer, is the first anti-angiogenesis drug delivered in combination with chemotherapy that has consistently shown clinical efficacy in the treatment of breast cancer. Since the ECOG 2100 trial has shown that bevacizumab added to paclitaxel as a first-line treatment for advanced breast cancer nearly doubled the time to progression and tumor response rate, its approval was granted almost worldwide. However, other phase III trials revealed a smaller absolute improvement in progression-free survival (PFS) and response rates, and no trials yet have demonstrated survival enhancement which led to a great controversy and debate over the use of bevacizumab. The discrepancy between PFS and overall survival also raises the question of whether or not bevacizumab has been applied sub-optimally in some of the studies, if a predictive biomarker(s) exists to select the group of patients whom would receive the greatest benefit and what is the appropriate clinical end-point for approval and funding of new targeted agents. In this article we will review the bevacizumab mechanism of action and the clinical trials that assessed its benefit in the treatment of metastatic breast cancer (MBC).
Collapse
Affiliation(s)
- Ana Elisa Lohmann
- British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
92
|
Hsieh YC, Jeng JS, Lin HJ, Hu CJ, Yu CC, Lien LM, Peng GS, Chen CI, Tang SC, Chi NF, Tseng HP, Chern CM, Hsieh FI, Bai CH, Chen YR, Chiou HY. Epistasis analysis for estrogen metabolic and signaling pathway genes on young ischemic stroke patients. PLoS One 2012; 7:e47773. [PMID: 23112845 PMCID: PMC3480403 DOI: 10.1371/journal.pone.0047773] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 09/17/2012] [Indexed: 12/19/2022] Open
Abstract
Background Endogenous estrogens play an important role in the overall cardiocirculatory system. However, there are no studies exploring the hormone metabolism and signaling pathway genes together on ischemic stroke, including sulfotransferase family 1E (SULT1E1), catechol-O-methyl-transferase (COMT), and estrogen receptor α (ESR1). Methods A case-control study was conducted on 305 young ischemic stroke subjects aged ≦ 50 years and 309 age-matched healthy controls. SULT1E1 -64G/A, COMT Val158Met, ESR1 c.454−397 T/C and c.454−351 A/G genes were genotyped and compared between cases and controls to identify single nucleotide polymorphisms associated with ischemic stroke susceptibility. Gene-gene interaction effects were analyzed using entropy-based multifactor dimensionality reduction (MDR), classification and regression tree (CART), and traditional multiple regression models. Results COMT Val158Met polymorphism showed a significant association with susceptibility of young ischemic stroke among females. There was a two-way interaction between SULT1E1 -64G/A and COMT Val158Met in both MDR and CART analysis. The logistic regression model also showed there was a significant interaction effect between SULT1E1 -64G/A and COMT Val158Met on ischemic stroke of the young (P for interaction = 0.0171). We further found that lower estradiol level could increase the risk of young ischemic stroke for those who carry either SULT1E1 or COMT risk genotypes, showing a significant interaction effect (P for interaction = 0.0174). Conclusions Our findings support that a significant epistasis effect exists among estrogen metabolic and signaling pathway genes and gene-environment interactions on young ischemic stroke subjects.
Collapse
Affiliation(s)
- Yi-Chen Hsieh
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Jiann-Shing Jeng
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Huey-Juan Lin
- Department of Neurology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Taipei Medical University Hospital and Shuang Ho Hospital, Taipei, Taiwan
| | - Chia-Chen Yu
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Li-Ming Lien
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Giia-Sheun Peng
- Department of Neurology, Tri-Service General Hospital, Taipei, Taiwan
| | - Chin-I Chen
- Department of Neurology, Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Sung-Chun Tang
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Nai-Fang Chi
- Department of Neurology, Taipei Medical University Hospital and Shuang Ho Hospital, Taipei, Taiwan
| | - Hung-Pin Tseng
- Department of Neurology, Lotung Poh-Ai Hospital, I-Lan, Taiwan
| | - Chang-Ming Chern
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Fang-I Hsieh
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Chyi-Huey Bai
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Yi-Rhu Chen
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yi Chiou
- School of Public Health, Taipei Medical University, Taipei, Taiwan
- Dr. Chi-Hsing Huang Stroke Research Center, Taipei Medical University, Taipei, Taiwan
- * E-mail:
| | | |
Collapse
|
93
|
Bernelot Moens SJ, Schnitzler GR, Nickerson M, Guo H, Ueda K, Lu Q, Aronovitz MJ, Nickerson H, Baur WE, Hansen U, Iyer LK, Karas RH. Rapid estrogen receptor signaling is essential for the protective effects of estrogen against vascular injury. Circulation 2012; 126:1993-2004. [PMID: 22997253 DOI: 10.1161/circulationaha.112.124529] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Clinical trial and epidemiological data support that the cardiovascular effects of estrogen are complex, including a mixture of both potentially beneficial and harmful effects. In animal models, estrogen protects females from vascular injury and inhibits atherosclerosis. These effects are mediated by estrogen receptors (ERs), which, when bound to estrogen, can bind to DNA to directly regulate transcription. ERs can also activate several cellular kinases by inducing a rapid nonnuclear signaling cascade. However, the biological significance of this rapid signaling pathway has been unclear. METHODS AND RESULTS In the present study, we develop a novel transgenic mouse in which rapid signaling is blocked by overexpression of a peptide that prevents ERs from interacting with the scaffold protein striatin (the disrupting peptide mouse). Microarray analysis of ex vivo treated mouse aortas demonstrates that rapid ER signaling plays an important role in estrogen-mediated gene regulatory responses. Disruption of ER-striatin interactions also eliminates the ability of estrogen to stimulate cultured endothelial cell migration and to inhibit cultured vascular smooth muscle cell growth. The importance of these findings is underscored by in vivo experiments demonstrating loss of estrogen-mediated protection against vascular injury in the disrupting peptide mouse after carotid artery wire injury. CONCLUSIONS Taken together, these results support the concept that rapid, nonnuclear ER signaling contributes to the transcriptional regulatory functions of ER and is essential for many of the vasoprotective effects of estrogen. These findings also identify the rapid ER signaling pathway as a potential target for the development of novel therapeutic agents.
Collapse
|
94
|
Barton M. Position paper: The membrane estrogen receptor GPER--Clues and questions. Steroids 2012; 77:935-42. [PMID: 22521564 DOI: 10.1016/j.steroids.2012.04.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 03/13/2012] [Accepted: 04/01/2012] [Indexed: 12/25/2022]
Abstract
Rapid signaling of estrogen involves membrane estrogen receptors (ERs), including membrane subpopulations of ERα and ERβ. In the mid-1990s, several laboratories independently reported the cloning of an orphan G protein-coupled receptor from vascular and cancer cells that was named GPR30. Research published between 2000 and 2005 provided evidence that GPR30 binds and signals via estrogen indicating that this intracellular receptor is involved in rapid, non-genomic estrogen signaling. The receptor has since been designated as the G protein-coupled estrogen receptor (GPER) by the International Union of Pharmacology. The availability of genetic tools such as different lines of GPER knock-out mice, as well as GPER-selective agonists and antagonists has advanced our understanding, but also added some confusion about the new function of this receptor. GPER not only binds estrogens but also other substances, including SERMs, SERDs, and environmental ER activators (endocrine disruptors; xenoestrogens) and also interacts with other proteins. This article represents a summary of a lecture given at the 7(th) International Meeting on Rapid Responses to Steroid Hormones in September 2011 in Axos, Crete, and reviews the current knowledge and questions about GPER-dependent signaling and function. Controversies that have complicated our understanding of GPER, including interactions with human ERα-36 and aldosterone as a potential ligand, will also be discussed.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zurich, LTK Y44 G22, Winterthurerstrasse 190, 8057 Zürich, Switzerland.
| |
Collapse
|
95
|
Chronic cranberry juice consumption restores cholesterol profiles and improves endothelial function in ovariectomized rats. Eur J Nutr 2012; 52:1145-55. [PMID: 22836513 DOI: 10.1007/s00394-012-0425-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 07/12/2012] [Indexed: 01/05/2023]
Abstract
PURPOSE Postmenopausal women experience higher risks for cardiovascular diseases than age-matched men and pre-menopausal women. There is a need for better treatment strategy for estrogen-deficient-related cardiovascular complications. We and others have recently reported that activated renin-angiotensin system and the associated oxidative stress impaired endothelium-dependent relaxation in ovariectomized rat, while angiotensin receptor blocker rescues endothelial dysfunction. Dietary supplements and lifestyle modifications provide an alternative way to improve cardiovascular health. The present study tests the hypothesis that chronic cranberry juice consumption improves cholesterol profiles and vascular functions in estrogen-deficient animal model. The effect of cranberry consumption on expression and activity of renin-angiotensin system in the vasculature will be determined. METHODS Ovariectomized rats were treated daily with commercial cranberry juice at 7 mg/kg for 8 weeks, a dosage comparable to recent clinical studies. Serum was collected for measuring cholesterol levels while aorta was isolated for isometric force assay and expression studies. RESULTS Cranberry juice consumption reduced circulating levels of total cholesterol, triacylglycerols, HDL, nHDL, and nHDL/HDL ratio. Meanwhile, cranberry juice consumption improved endothelium-dependent relaxation in aorta of ovariectomized rats by restoring p-eNOS level (endothelial nitric oxide synthase phosphorylated at ser-1177), reversing the up-regulated levels of renin-angiotensin system markers (angiotensin-converting enzyme, angiotensin II, and angiotensin II type 1 receptor), and normalizing the elevated NAD(P)H oxidase expression and oxidative stress. CONCLUSIONS Our data demonstrate the novel cardiovascular benefits of cranberry juice consumption in improving both vascular functions and cholesterol profiles, providing insight into developing cranberry products into useful dietary supplements for postmenopausal women.
Collapse
|
96
|
Thomas GW, Rael LT, Bar-Or R, Mains CW, Slone DS, Boyd SR, Bar-Or D. Biphasic effect of danazol on human vascular endothelial cell permeability and f-actin cytoskeleton dynamics. Biochem Biophys Res Commun 2012; 421:707-12. [PMID: 22542943 DOI: 10.1016/j.bbrc.2012.04.066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 04/13/2012] [Indexed: 10/28/2022]
Abstract
Breakdown of endothelial barrier function is a hallmark event across a variety of pathologies such as inflammation, cancer, and diabetes. It has also been appreciated that steroid hormones impart direct biological activity on endothelial cells at many levels. The purpose of this investigation was to explore the effect of the androgen-like steroid, danazol, on endothelial cell barrier function in vitro. Primary human endothelial cells exposed to 0.01-50 μM danazol were evaluated for changes in permeability. We found that danazol altered endothelial permeability in a biphasic manner in which nanomolar concentrations enhance barrier function while micromolar concentrations are detrimental. Monitoring of trans-endothelial electrical resistance demonstrated that these barrier enhancing effects were rapid (within 5 min) and lasted for over 24h. Analysis of intracellular f-actin organization showed that barrier enhancement also correlated with the formation of a submembranous cortical actin ring. Conversely, at higher danazol concentrations, contractile cell phenotypes were observed, represented by stress fiber formation. Competitive binding studies performed using steroid hormone receptor antagonists proved that this activity is the result of androgen and estrogen receptor ligation. These findings suggest that low dose danazol may provide a therapeutic window for diseases involving vascular leakage.
Collapse
Affiliation(s)
- Gregory W Thomas
- Swedish Medical Center, Trauma Research Laboratory, Englewood, CO, USA
| | | | | | | | | | | | | |
Collapse
|
97
|
Velnar T, Smrkolj V, Rupnik MS, Gradisnik L. Is tissue augmentation a reality in biosurgery? An experimental study of endothelial cell invasion into tissue filler. Int Wound J 2012; 10:321-8. [PMID: 22487593 DOI: 10.1111/j.1742-481x.2012.00980.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
New therapeutic approaches for wound treatment are evolving. Non healing wounds in oncology and after trauma may be cured by a novel technique of tissue augmentation with soft tissue fillers. The principle resides in filling the wound with collagen filler in order to seal the defect and promote healing. Successful angiogenesis forms the basis of tissue filler survival and determines the outcome of the healing process. During this study, basic data about endothelial cell invasion into collagen-made substratum was collected that could be used for neoangiogenesis studies in tissue augmentation techniques for large wound defect treatment. In the in vitro assay, the human umbilical vein endothelial cells (HUVEC) grow into a three-dimensional framework of collagenous tissue fillers, forming the basic step for angiogenesis. After heparins were used as chemotactic agents, a typical bell-shaped relationship between chemotaxis and agent concentrations was found. Significant cell infiltration was present in the assays with chemotactic agents. These observations support the potential for tissue augmentation with soft tissue fillers that could be used in acute and chronic non healing traumatic and oncology wounds after extensive surgical resections and radiotherapy.
Collapse
Affiliation(s)
- Tomaz Velnar
- Department of Neurosurgery, University Medical Centre Ljubljana, Slovenia Laboratory Centre of Medical Faculty Maribor, Maribor, Slovenia.
| | | | | | | |
Collapse
|
98
|
Pastore MB, Jobe SO, Ramadoss J, Magness RR. Estrogen receptor-α and estrogen receptor-β in the uterine vascular endothelium during pregnancy: functional implications for regulating uterine blood flow. Semin Reprod Med 2012; 30:46-61. [PMID: 22271294 DOI: 10.1055/s-0031-1299597] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The steroid hormone estrogen and its classical estrogen receptors (ERs), ER-α and ER-β, have been shown to be partly responsible for the short- and long-term uterine endothelial adaptations during pregnancy. The ER-subtype molecular and structural differences coupled with the differential effects of estrogen in target cells and tissues suggest a substantial functional heterogeneity of the ERs in estrogen signaling. In this review we discuss (1) the role of estrogen and ERs in cardiovascular adaptations during pregnancy, (2) in vivo and in vitro expression of ERs in uterine artery endothelium during the ovarian cycle and pregnancy, contrasting reproductive and nonreproductive arterial endothelia, (3) the structural basis for functional diversity of the ERs and estrogen subtype selectivity, (4) the role of estrogen and ERs on genomic responses of uterine artery endothelial cells, and (5) the role of estrogen and ERs on nongenomic responses in uterine artery endothelia. These topics integrate current knowledge of this very rapidly expanding scientific field with diverse interpretations and hypotheses regarding the estrogenic effects that are mediated by either or both ERs and their relationship with vasodilatory and angiogenic vascular adaptations required for modulating the dramatic physiological rises in uteroplacental perfusion observed during normal pregnancy.
Collapse
Affiliation(s)
- Mayra B Pastore
- Department of Obstetrics/Gynecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | |
Collapse
|
99
|
Matsubara Y, Matsubara K. Estrogen and progesterone play pivotal roles in endothelial progenitor cell proliferation. Reprod Biol Endocrinol 2012; 10:2. [PMID: 22252173 PMCID: PMC3395836 DOI: 10.1186/1477-7827-10-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 01/17/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND It has been previously suggested that angiogenesis occurs during the menstrual cycle. Moreover, a rise in uterine blood flow is largely maintained by vasodilatation and substantial increases in angiogenesis. It is known that estradiol (E2) and progesterone (P4) are involved in angiogenesis. Recently, endothelial progenitor cells (EPCs) were found to be involved in neovascularization; however, their roles in uterine neovascularization have not been well characterized. We hypothesized that E2- or P4-mediated EPC proliferation plays important roles in uterine neovascularization during the menstrual cycle. METHODS The number of EPCs in peripheral blood from subjects in the menstrual phase (n=12), follicular phase (n=8), and luteal phase (n=16), was measured using flow cytometry. Peripheral blood mononuclear cells (PBMCs) were cultured for seven days with or without 17beta-estradiol (E2beta) or P4, followed by assessment of EPC proliferation based upon the uptake of acetylated low density lipoprotein (LDL) and lectin. The expression of estrogen receptor (ER) or progesterone receptor (PR) in EPCs was also evaluated using real-time PCR. RESULTS E2beta and P4 significantly increased the proliferation of EPCs derived from the peripheral blood of subjects in menstrual phase, but not subjects in the luteal phase. In addition, the expression level of ERalpha was markedly higher than ERbeta in EPCs derived from women in menstrual phase. CONCLUSIONS EPC proliferation is induced during the menstrual phase and proliferation can be affected by estrogen through ERalpha activation.
Collapse
Affiliation(s)
- Yuko Matsubara
- Department of Obstetrics and Gynecology, Ehime Prefectural Niihama Hospital, Hongo, Niihama, Ehime, 792-0042 Japan
| | - Keiichi Matsubara
- Department of Obstetrics and Gynecology, Ehime Prefectural Niihama Hospital, Hongo, Niihama, Ehime, 792-0042 Japan
| |
Collapse
|
100
|
Straub RH, Cutolo M, Fleck M. Rheumatoid Arthritis Recapitulates Events Relevant in Blastocyst Implantation and Embryogenesis: A Pathogenetic Theory. Semin Arthritis Rheum 2011; 41:382-92. [DOI: 10.1016/j.semarthrit.2011.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 04/05/2011] [Accepted: 04/18/2011] [Indexed: 01/07/2023]
|