51
|
Hypoxia: Turning vessels into vassals of cancer immunotolerance. Cancer Lett 2020; 487:74-84. [PMID: 32470491 DOI: 10.1016/j.canlet.2020.05.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/27/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
Hypoxia is a universal feature of solid cancers caused by a mismatch between cellular oxygen supply and consumption. To meet the increased demand for oxygen, hypoxic cancer cells (CCs) induce a multifaceted process known as angiogenesis, wherein new vessels are formed by the sprouting of pre-existing ones. In addition to providing oxygen for growth and an exit route for dissemination, angiogenic vessels and factors are co-opted by CCs to enable the generation of an immunotolerant, hypoxic tumor microenvironment, leading to therapeutic failure and mortality. In this review, we discuss how hypoxia-inducible factors (HIFs), the mechanistic target of rapamycin (mTOR), and the unfolded protein response (UPR) control angiogenic factors serving both vascular and immunomodulatory functions in the tumor microenvironment. Possible therapeutic strategies, wherein targeting oxygen sensing might enhance anti-angiogenic and immunologically-mediated anti-cancer responses, are suggested.
Collapse
|
52
|
Zhou B, Ge T, Zhou L, Jiang L, Zhu L, Yao P, Yu Q. Dimethyloxalyl Glycine Regulates the HIF-1 Signaling Pathway in Mesenchymal Stem Cells. Stem Cell Rev Rep 2020; 16:702-710. [DOI: 10.1007/s12015-019-09947-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
53
|
Rajakumar A, Kane MA, Yu J, Taylor RN, Sidell N. Aberrant retinoic acid production in the decidua: Implications for pre-eclampsia. J Obstet Gynaecol Res 2020; 46:1007-1016. [PMID: 32343034 DOI: 10.1111/jog.14262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/03/2020] [Indexed: 01/05/2023]
Abstract
Fine-tuning of the endometrium during the evanescent 'window of implantation' relies upon an array of diverse and redundant signaling molecules, particularly the ovarian steroids E2 and P4, but also growth factors, eicosanoids, and vitamins including the vitamin A compounds (retinoids). Pregnancy complications such as preeclampsia (PE) can result from aberrations in the production or function of these molecules that arise during this critical period of decidual development. Such aberrations may be reflected by incomplete decidualization, reduced spiral artery modification, and/or loss of immune tolerance to the developing fetus. Our understanding of the role of the active retinoid metabolite all-trans retinoic acid (RA) in maintaining immune balance in certain tissues, along with data describing its role in decidualization, present a compelling argument that aberrant RA signaling in the decidua can play a significant role in the etiology of PE. Recent findings that decidualization and expression of the anti-angiogenic gene product, 'soluble fms-like tyrosine kinase-1' (sFLT1) are negatively correlated and that sFLT1 expression is directly inhibited by RA, provide additional evidence of the critical role of this retinoid in regulating early vascular development in the decidua. This review provides insight into the production and function of RA in the decidua and how modifications in its metabolism and signaling might lead to certain pregnancy disorders such as PE.
Collapse
Affiliation(s)
- Augustine Rajakumar
- Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Jie Yu
- Department of Obstetrics & Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Robert N Taylor
- Department of Obstetrics & Gynecology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Neil Sidell
- Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
54
|
Zhou C, Zou QY, Jiang YZ, Zheng J. Role of oxygen in fetoplacental endothelial responses: hypoxia, physiological normoxia, or hyperoxia? Am J Physiol Cell Physiol 2020; 318:C943-C953. [PMID: 32267717 DOI: 10.1152/ajpcell.00528.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
During pregnancy, placental vascular growth, which is essential for supporting the rapidly growing fetus, is associated with marked elevations in blood flow. These vascular changes take place under chronic physiological low O2 (less than 2-8% O2 in human; chronic physiological normoxia, CPN) throughout pregnancy. O2 level below CPN pertinent to the placenta results in placental hypoxia. Such hypoxia can cause severe endothelial dysfunction, which is associated with adverse pregnancy outcomes (e.g., preeclampsia) and high risk of adult-onset cardiovascular diseases in children born to these pregnancy complications. However, our current knowledge about the mechanisms underlying fetoplacental endothelial function is derived primarily from cell models established under atmospheric O2 (~21% O2 at sea level, hyperoxia). Recent evidence has shown that fetoplacental endothelial cells cultured under CPN have distinct gene expression profiles and cellular responses compared with cells cultured under chronic hyperoxia. These data indicate the critical roles of CPN in programming fetal endothelial function and prompt us to re-examine the mechanisms governing fetoplacental endothelial function under CPN. Better understanding these mechanisms will facilitate us to develop preventive and therapeutic strategies for endothelial dysfunction-associated diseases (e.g., preeclampsia). This review will provide a brief summary on the impacts of CPN on endothelial function and its underlying mechanisms with a focus on fetoplacental endothelial cells.
Collapse
Affiliation(s)
- Chi Zhou
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Qing-Yun Zou
- Department of Vascular Surgery, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yi-Zhou Jiang
- Institute for Advanced Study, Shenzhen University, Shenzhen, Guangdong, China
| | - Jing Zheng
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin.,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
55
|
Mammadzada P, Corredoira PM, André H. The role of hypoxia-inducible factors in neovascular age-related macular degeneration: a gene therapy perspective. Cell Mol Life Sci 2020; 77:819-833. [PMID: 31893312 PMCID: PMC7058677 DOI: 10.1007/s00018-019-03422-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022]
Abstract
Understanding the mechanisms that underlie age-related macular degeneration (AMD) has led to the identification of key molecules. Hypoxia-inducible transcription factors (HIFs) have been associated with choroidal neovascularization and the progression of AMD into the neovascular clinical phenotype (nAMD). HIFs regulate the expression of multiple growth factors and cytokines involved in angiogenesis and inflammation, hallmarks of nAMD. This knowledge has propelled the development of a new group of therapeutic strategies focused on gene therapy. The present review provides an update on current gene therapies in ocular angiogenesis, particularly nAMD, from both basic and clinical perspectives.
Collapse
Affiliation(s)
- Parviz Mammadzada
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden
| | - Pablo M Corredoira
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden
| | - Helder André
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden.
| |
Collapse
|
56
|
The Pathogenesis of Adenomyosis vis-à-vis Endometriosis. J Clin Med 2020; 9:jcm9020485. [PMID: 32050720 PMCID: PMC7073526 DOI: 10.3390/jcm9020485] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/31/2020] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
Adenomyosis is used to be called endometriosis interna, and deep endometriosis is now called adenomyosis externa. Thus, there is a question as to whether adenomyosis is simply endometriosis of the uterus, either from the perspective of pathogenesis or pathophysiology. In this manuscript, a comprehensive review was performed with a literature search using PubMed for all publications in English, related to adenomyosis and endometriosis, from inception to June 20, 2019. In addition, two prevailing theories, i.e., invagination—based on tissue injury and repair (TIAR) hypothesis—and metaplasia, on adenomyosis pathogenesis, are briefly overviewed and then critically scrutinized. Both theories have apparent limitations, i.e., difficulty in falsification, explaining existing data, and making useful predictions. Based on the current understanding of wound healing, a new hypothesis, called endometrial-myometrial interface disruption (EMID), is proposed to account for adenomyosis resulting from iatrogenic trauma to EMI. The EMID hypothesis not only highlights the more salient feature, i.e., hypoxia, at the wounding site, but also incorporates epithelial mesenchymal transition, recruitment of bone-marrow-derived stem cells, and enhanced survival and dissemination of endometrial cells dispersed and displaced due to iatrogenic procedures. More importantly, the EMID hypothesis predicts that the risk of adenomyosis can be reduced if certain perioperative interventions are performed. Consequently, from a pathogenic standpoint, adenomyosis is not simply endometriosis of the uterus, and, as such, may call for interventional procedures that are somewhat different from those for endometriosis to achieve the best results.
Collapse
|
57
|
Sasagawa T, Jinno-Oue A, Nagamatsu T, Morita K, Tsuruga T, Mori-Uchino M, Fujii T, Shibuya M. Production of an anti-angiogenic factor sFLT1 is suppressed via promoter hypermethylation of FLT1 gene in choriocarcinoma cells. BMC Cancer 2020; 20:112. [PMID: 32041578 PMCID: PMC7011436 DOI: 10.1186/s12885-020-6598-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/03/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Soluble Fms-like tyrosine kinase-1 (sFLT1) as an anti-angiogenic factor is abundantly expressed in placental trophoblasts. Choriocarcinoma, a malignant tumor derived from trophoblasts, is known to be highly angiogenic and metastatic. However, the molecular mechanism underlying angiogenesis in choriocarcinoma pathogenesis remains unclear. We aimed to investigate the mRNA expression and DNA methylation status of the FLT1 gene in human choriocarcinoma cells and trophoblast cells. METHODS qRT-PCR, Western blotting and ELISA were conducted to evaluate the mRNA and protein expression levels of sFLT1. 5-aza-2'-deoxycytidine (5azadC) treatment and bisulfite sequencing were used to study the FLT1 gene promoter methylation. The effect of sFLT1 on choriocarcinoma growth and angiogenesis was evaluated in a xenograft mouse model. RESULTS Expression of the FLT1 gene was strongly suppressed in choriocarcinoma cell lines compared with that in the primary trophoblasts. Treatment of choriocarcinoma cell lines with 5azadC, a DNA methyltransferase inhibitor, markedly increased in mRNA expression of three FLT1 splice variants and secretion of sFLT1 proteins. Bisulfite sequencing revealed that the CpG hypermethylation was observed at the FLT1 promoter region in choriocarcinoma cell lines and a human primary choriocarcinoma tissue but not in human trophoblast cells. Interestingly, in 5azadC-treated choriocarcinoma cell lines, sFLT1 mRNA expression and sFLT1 production were further elevated by hypoxic stimulation. Finally, as expected, sFLT1-expressing choriocarcinoma cells implanted into nude mice showed significantly slower tumor growth and reduced microvessel formation compared with GFP-expressing control choriocarcinoma cells. CONCLUSIONS Inhibition of sFLT1 production by FLT1 silencing occurs via the hypermethylation of its promoter in choriocarcinoma cells. The stable expression of sFLT1 in choriocarcinoma cells resulted in the suppression of tumor growth and tumor vascularization in vivo. We suggest that the FLT1 gene may be a cell-type-specific tumor suppressor in choriocarcinoma cells.
Collapse
Affiliation(s)
- Tadashi Sasagawa
- Institute of Physiology and Medicine, Jobu University, 270-1 Shin-machi, Takasaki, Gunma, 370-1393, Japan
| | - Atsushi Jinno-Oue
- Bioresource Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazuki Morita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tetsushi Tsuruga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Mayuyo Mori-Uchino
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masabumi Shibuya
- Institute of Physiology and Medicine, Jobu University, 270-1 Shin-machi, Takasaki, Gunma, 370-1393, Japan.
| |
Collapse
|
58
|
Iriyama T, Wang W, Parchim NF, Sayama S, Kumasawa K, Nagamatsu T, Song A, Xia Y, Kellems RE. Reciprocal upregulation of hypoxia-inducible factor-1α and persistently enhanced placental adenosine signaling contribute to the pathogenesis of preeclampsia. FASEB J 2020; 34:4041-4054. [PMID: 31930569 DOI: 10.1096/fj.201902583r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/28/2019] [Accepted: 12/30/2019] [Indexed: 02/02/2023]
Abstract
Recent evidence indicates that elevated placental adenosine signaling contributes to preeclampsia (PE). However, the molecular basis for the chronically enhanced placental adenosine signaling in PE remains unclear. Here, we report that hypoxia-inducible factor-1α (HIF-1α) is crucial for the enhancement of placental adenosine signaling. Utilizing a pharmacologic approach to reduce placental adenosine levels, we found that enhanced adenosine underlies increased placental HIF-1α in an angiotensin receptor type 1 receptor agonistic autoantibody (AT1 -AA)-induced mouse model of PE. Knockdown of placental HIF-1α in vivo suppressed the accumulation of adenosine and increased ecto-5'-nucleotidase (CD73) and adenosine A2B receptor (ADORA2B) in the placentas of PE mouse models induced by AT1 -AA or LIGHT, a TNF superfamily cytokine (TNFSF14). Human in vitro studies using placental villous explants demonstrated that increased HIF-1α resulting from ADORA2B activation facilitates the induction of CD73, ADORA2B, and FLT-1 expression. Overall, we demonstrated that (a) elevated placental HIF-1α by AT1 -AA or LIGHT upregulates CD73 and ADORA2B expression and (b) enhanced adenosine signaling through upregulated ADORA2B induces placental HIF-1α expression, which creates a positive feedback loop that promotes FLT-1 expression leading to disease development. Our results suggest that adenosine-based therapy targeting the malicious cycle of placental adenosine signaling may elicit therapeutic effects on PE.
Collapse
Affiliation(s)
- Takayuki Iriyama
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Wei Wang
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA.,Department of Nephrology, Xiangya Hospital of Central South University, Changsha, P.R. China
| | - Nicholas F Parchim
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA.,Department of Emergency Medicine, The University of New Mexico Hospital, Albuquerque, NM, USA
| | - Seisuke Sayama
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Keiichi Kumasawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Anren Song
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA.,Department of Nephrology, Xiangya Hospital of Central South University, Changsha, P.R. China.,Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rodney E Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
59
|
Zhu J, Zhang J, Ng MJ, Chern B, Yeo GS, Tan KH. Angiogenic factors during pregnancy in Asian women with elevated blood pressure in early pregnancy and the risk of preeclampsia: a longitudinal cohort study. BMJ Open 2019; 9:e032237. [PMID: 31791965 PMCID: PMC6924722 DOI: 10.1136/bmjopen-2019-032237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE It remains unclear what roles placenta-originated angiogenic factors play in the pathogenesis of preeclampsia among hypertensive women. We compared maternal soluble fms-like tyrosine kinase 1 (sFlt-1) and placental growth factor (PlGF) levels throughout pregnancy in women with normal blood pressure (BP), elevated BP and hypertension in early pregnancy and their risks of developing preeclampsia. DESIGN A prospective cohort study. SETTING KK Women's and Children's Hospital, Singapore. PARTICIPANTS 923 women with singleton pregnancy <14 weeks of gestation were included in the prospective Neonatal and Obstetrics Risks Assessment cohort between September 2010 and October 2014. Systolic, diastolic, mean arterial blood pressure (MAP) were measured at 11-14 weeks. PRIMARY AND SECONDARY OUTCOMES Maternal serum sFlt-1, PlGF and sFlt-1/PlGF ratio were tested at 11-14, 18-22, 28-32 and 34 weeks onwards of gestation. Preeclampsia was main pregnancy outcome. RESULTS Women were divided based on their BP in early pregnancy: normal (n=750), elevated BP (n=98) and hypertension (n=75). Maternal sFlt-1 levels and sFlt-1/PlGF ratios were higher in hypertensive women throughout pregnancy, but maternal PlGF levels were not significantly lower. Rise in maternal systolic, diastolic BP and MAP at 11-14 weeks were significantly associated with higher sFlt-1/PlGF ratios during pregnancy. A 10 mm Hg increase in MAP was associated with a 5.6-fold increase in risk of preterm preeclampsia and a 3.3-fold increase in risk of term preeclampsia, respectively. CONCLUSION Women with elevated BP in early pregnancy already had a higher sFlt-1/PlGF ratio in early gestation and throughout pregnancy, and an increased risk of preeclampsia. In contrast, PlGF levels in these women remained normal.
Collapse
Affiliation(s)
- Jing Zhu
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Division of Obstetrics and Gynaecology, KK Women's and Children's Hospital, Singapore, Singapore
| | - Mor Jack Ng
- Division of Obstetrics and Gynaecology, KK Women's and Children's Hospital, Singapore, Singapore
| | - Bernard Chern
- Division of Obstetrics and Gynaecology, KK Women's and Children's Hospital, Singapore, Singapore
| | - George Sh Yeo
- Department of Maternal and Fetal Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Kok Hian Tan
- Division of Obstetrics and Gynaecology, KK Women's and Children's Hospital, Singapore, Singapore
| |
Collapse
|
60
|
Genetic variants of VEGFR-1 gene promoter in acute myocardial infarction. Hum Genomics 2019; 13:56. [PMID: 31744542 PMCID: PMC6862733 DOI: 10.1186/s40246-019-0243-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/09/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Coronary artery disease (CAD) including acute myocardial infarction (AMI) is a common complex disease caused by atherosclerosis. Vascular epithelial growth factor receptor-1 (VEGFR-1) stimulates angiogenesis and vascular permeability, and functions as a decoy to sequester VEGF and prevent initiation of intracellular signaling. VEGFR-1 knockout mice exhibit significantly higher mortality due to heart failure, cardiac hypertrophy, and cardiac dysfunction. An evident increase in macrophage infiltration and cardiac fibrosis are also observed after transverse aortic constriction. Therefore, VEGFR-1 gene variants may be involved in CAD. In this study, VEGFR-1 gene promoter was genetically and functionally analyzed in large cohorts of AMI patients and ethnic-matched controls. RESULTS A total of 16 DNA sequence variants (DSVs) including six single-nucleotide polymorphisms (SNPs) were found in the VEGFR-1 gene promoter and 5'-untranslated region. Five novel DSVs and one SNP were only identified in AMI patients group. These DSVs and SNP significantly altered the transcriptional activity of the VEGFR-1 gene promoter in both HEK-293 and H9c2 cells (P < 0.05). Further electrophoretic mobility shift assay indicated that the DSVs and SNPs evidently affected the binding of transcription factors. CONCLUSIONS The genetic variants in VEGFR-1 gene identified in AMI patients may alter the transcriptional activity of the VEGFR-1 gene promoter and change VEGFR-1 level, contributing to AMI development.
Collapse
|
61
|
Deepak V, Sahu MB, Yu J, Jones JW, Kane MA, Taylor RN, Badell ML, Sidell N, Rajakumar A. Retinoic Acid Is a Negative Regulator of sFLT1 Expression in Decidual Stromal Cells, and Its Levels Are Reduced in Preeclamptic Decidua. Hypertension 2019; 73:1104-1111. [PMID: 30879360 DOI: 10.1161/hypertensionaha.118.12564] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
sFLT1 (soluble VEGF [vascular endothelial growth factor] receptor-1) levels are increased in preeclampsia-a pathological condition of pregnancy. The mechanism of sFLT1 overexpression by gestational tissues, particularly the decidua, remains unknown. Mass spectrometry measurement of the active retinoid metabolite, all-trans retinoic acid (RA), showed significantly lower levels of RA in preeclamptic versus normotensive decidua. In this study, we investigated the involvement of RA in regulating decidual sFLT1 expression. When decidual stromal cells (DSCs) isolated from the decidua basalis of normotensive and preeclampsia placentas were treated with BMS493-a pan-RAR (RA nuclear receptor) antagonist-upregulation of sFLT1 expression was observed. Conversely, treatment with RA resulted in downregulation of sFLT1 in normotensive DSCs and preeclampsia DSCs. Unlike treatment with cAMP, which induces decidualization while downregulating sFLT1, RA treatment did not alter DSC expression of prolactin-a marker of decidualization-or FOXO1 (forkhead box protein 01)-a transcription factor required for prolactin upregulation. TFAP2A (transcription factor AP-2-alpha [activating enhancer-binding protein 2 alpha]), a different transcription factor was upregulated in normotensive DSCs but not in preeclampsia DSCs after RA treatment. Collectively, our data show that RA suppresses sFLT1 expression in DSCs independently of cellular decidualization. These findings suggest that reduced decidual RA levels may contribute to preeclampsia pathogenesis by allowing sFLT1 accumulation at the maternal-fetal interface.
Collapse
Affiliation(s)
- Venkataraman Deepak
- From the Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA (V.D., M.B.S., M.L.B., N.S., A.R.)
| | - Margaret B Sahu
- From the Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA (V.D., M.B.S., M.L.B., N.S., A.R.)
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore (J.Y., J.W.J., M.A.K.)
| | - Jace W Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore (J.Y., J.W.J., M.A.K.)
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore (J.Y., J.W.J., M.A.K.)
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City (R.N.T.)
| | - Martina L Badell
- From the Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA (V.D., M.B.S., M.L.B., N.S., A.R.)
| | - Neil Sidell
- From the Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA (V.D., M.B.S., M.L.B., N.S., A.R.)
| | - Augustine Rajakumar
- From the Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA (V.D., M.B.S., M.L.B., N.S., A.R.)
| |
Collapse
|
62
|
Effect of magnesium-degradation products and hypoxia on the angiogenesis of human umbilical vein endothelial cells. Acta Biomater 2019; 98:269-283. [PMID: 30794987 DOI: 10.1016/j.actbio.2019.02.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 01/06/2023]
Abstract
Biodegradable magnesium (Mg) metals have been applied in orthopaedic and stent applications due to their biodegradability, bioabsorbability and adaptability to tissue regeneration. However, further investigations are still needed to understand how angiogenesis will respond to high concentrations of Mg and oxygen content differences, which are vital to vascular remodelling and bone fracture regeneration or tissue healing. Human primary endothelial cells were exposed to various concentrations (2-8 mM) of extracellular Mg degradation products under either hypoxia or normoxia. Increased proliferation was measured with Mg extracts under hypoxia but not under normoxia. Under normoxia and with Mg extracts, HUVEC migration exhibited a bell-shaped curve. The same pattern was observed with VEGFB expression, while VEGFA was constantly downregulated. Under hypoxia, migration and VEGFA levels remained constant; however, VEGFB was upregulated. Similarly, under normoxia, tube formation as well as VEGFA and VEGFB levels were downregulated. Nevertheless, under hypoxia, tube formation remained constant while VEGFA and VEGFB levels were upregulated. These results suggest that Mg extracts did not interfere with angiogenesis under hypoxia. STATEMENT OF SIGNIFICANCE: Neoangiogenesis, mediated by (e.g.) hypoxia, is a key factor for proper tissue healing Thus, effect of Mg degradation products under either hypoxia or normoxia on angiogenesis were investigated. Under normoxia and increased Mg concentrations, a general negative effect was measured on early (migration) and late (tubulogenesis) angiogenesis. However, under hypoxia, this effect was abolished. As magnesium degradation is an oxygen-dependant process, hypoxia condition may be a relevant factor to test material cytocompatibility in vitro.
Collapse
|
63
|
Allogeneic Adipose-Derived Mesenchymal Stem Cell Transplantation Enhances the Expression of Angiogenic Factors in a Mouse Acute Hindlimb Ischemic Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1083:1-17. [PMID: 28687961 DOI: 10.1007/5584_2017_63] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell migration and molecular mechanisms during healing of damaged vascular or muscle tissues are emerging fields of interest worldwide. The study herein focuses on evaluating the role of allogenic adipose-derived mesenchymal stem cells (ADMSCs) in restoring damaged tissues. Using a hindlimb ischemic mouse model, ADMSC-mediated induction of cell migration and gene expression related to myocyte regeneration and angiogenesis were evaluated. ADMSCs were labeled with GFP (ADMSC-GFP). The proximal end of the femoral blood vessel of mice (over 6 months of age) are ligated at two positions then cut between the two ties. Hindlimb ischemic mice were randomly divided into two groups: Group I (n = 30) which was injected with PBS (100 μL) and Group II (n = 30) which was transplanted with ADMSC-GFP (106 cells/100 μL PBS) at the rectus femoris muscle. The migration of ADMSC-GFP in hindlimb was analyzed by UV-Vis system. The expression of genes related to angiogenesis and muscle tissue repair was quantified by real-time RT-PCR. The results showed that ADMSCs existed in the grafted hindlimb for 7 days. Grafted cells migrated to other damaged areas such as thigh and heel. In both groups the ischemic hindlimb showed an increased expression of several angiogenic genes, including Flt-1, Flk-1, and Ang-2. In particular, the expression of Ang-2 and myogenic-related gene MyoD was significantly increased in the ADMSC-treated group compared to the PBS-treated (control) group; the expression increased at day 28 compared to day 3. The other factors, such as VE-Cadherin, HGF, CD31, Myf5, and TGF-β, were also more highly expressed in the ADMSC-treated group than in the control group. Thus, grafted ADMSCs were able to migrate to other areas in the injured hindlimb, persist for approximately 7 days, and have a significantly positive impact on stimulating expression of myogenic- and angiogenesis-related genes.
Collapse
|
64
|
Zhan K, Bai L, Wang G, Zuo B, Xie L, Wang X. Different angiogenesis modes and endothelial responses in implanted porous biomaterials. Integr Biol (Camb) 2019; 10:406-418. [PMID: 29951652 DOI: 10.1039/c8ib00061a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An in vivo experimental model based on implanting porous biomaterials to study angiogenesis was proposed. In the implanted porous polyvinyl alcohol, three major modes of angiogenesis, sprouting, intussusception and splitting, were found. By electron microscopy and three-dimensional simulation of the angiogenic vessels, we investigated the morphological characteristics of the three modes and paid special attention to the initial morphological difference between intussusception and splitting, and it was confirmed that the endothelial abluminal invagination and intraluminal protrusion are pre-representations of intussusception and splitting, respectively. Based on immunohistochemical analysis of HIF-1α, VEGF and Flt-1 expressions, it was demonstrated that the dominant mode of angiogenesis is related to the local hypoxic condition, and that there is difference in the response of endothelial cells to hypoxia-induced VEGF between sprouting and splitting. Specifically, in the biomaterials implanted for 3 days, the higher expression and gradient of VEGF induced by severe hypoxia in the avascular area caused sprouting of the peripheral capillaries, and in the biomaterial implanted for 9 days, with moderate hypoxia, splitting became a dominant mode. Whether on day 3 or day 9, Flt-1 expression in sprouting endothelia was significantly higher than that in splitting endothelia, which indicates that sprouting is caused by the strong response of endothelial cells to VEGF, while splitting is associated with their weaker response. As a typical experimental example, these results show the effectiveness of the porous biomaterial implantation model for studying angiogenesis, which is expected to become a new general model.
Collapse
Affiliation(s)
- Kuihua Zhan
- School of Mechanical and Electric Engineering, Soochow University, 8 Jixue Road, Suzhou, 215131, China.
| | | | | | | | | | | |
Collapse
|
65
|
Lin L, Li G, Zhang W, Wang YL, Yang H. Low-dose aspirin reduces hypoxia-induced sFlt1 release via the JNK/AP-1 pathway in human trophoblast and endothelial cells. J Cell Physiol 2019; 234:18928-18941. [PMID: 31004367 DOI: 10.1002/jcp.28533] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 11/06/2022]
Abstract
Pre-eclampsia (PE) is a serious hypertensive disorder of pregnancy that remains a leading cause of perinatal and maternal morbidity and mortality worldwide. Placental ischemia/hypoxia and the secretion of soluble fms-like tyrosine kinase 1 (sFlt1) into maternal circulation are involved in the pathogenesis of PE. Although low-dose aspirin (LDA) has beneficial effects on the prevention of PE, the exact mechanisms of action of LDA, particularly on placental dysfunction, and sFlt1 release, have not been well investigated. This study aimed to determine whether LDA exists the protective effects on placental trophoblast and endothelial functions and prevents PE-associated sFlt1 release. First, we observed that LDA mitigated hypoxia-induced trophoblast apoptosis, showed positive effects on trophoblast cells migration and invasion activity, and increased the tube-forming activity of human umbilical vein endothelial cells (HUVECs). In addition, LDA decreased hypoxia-induced sFlt1 production, and the c-Jun NH2 -terminal kinase/activator protein-1 (JNK/AP-1) pathway was shown to mediate the induction of sFlt1. Moreover, the transcription factor AP-1 was confirmed to regulate the Flt1 gene expression by directly binding to the Flt1 promoter in luciferase assays. The result of chromatin immunoprecipitation assays further demonstrated that LDA could directly decrease the expression of the transcription factor AP-1, and thus decrease sFlt1 production. Finally, the effects of LDA on sFlt1 production were proved in human placental explants. Taken together, our data show the protective effects of LDA against trophoblast and endothelial cell dysfunction and reveal that the LDA-mediated inhibition of sFlt1 via the JNK/AP-1 pathway may be a potential cellular/molecular mechanism for the prevention of PE.
Collapse
Affiliation(s)
- Li Lin
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Guanlin Li
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Wanyi Zhang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing, China
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Maternal-Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| |
Collapse
|
66
|
Lee HR, Pelaez F, Silbaugh AM, Leslie F, Racila E, Azarin SM. Biomaterial Platform To Establish a Hypoxic Metastatic Niche in Vivo. ACS APPLIED BIO MATERIALS 2019; 2:1549-1560. [PMID: 35026889 DOI: 10.1021/acsabm.8b00837] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hypoxia is a hallmark of tumor microenvironments, exerting wide-ranging impacts on key processes of tumor progression and metastasis. However, our understanding of how hypoxia regulates these processes has been based primarily on studying the effects of hypoxia within the primary tumor. Recently, an increasing number of studies have suggested the importance of hypoxic regulation within metastatic target organs, but hypoxic metastatic niches in the body are difficult to access with current imaging techniques, hampering detailed in vivo investigation of hypoxia at metastatic sites. Here, we report an engineered biomaterial scaffold that is able to establish an in vivo hypoxic metastatic niche in a readily accessible area, enabling the investigation of hypoxic regulation at a metastatic site. We engineered hypoxic environments within microporous poly(lactide-co-glycolide) (PLG) scaffolds, which have previously been shown to act as premetastatic niche mimics, via the addition of CoCl2, a hypoxia-mimetic agent. When implanted into the subcutaneous region of mice, CoCl2-containing PLG (Co-PLG) scaffolds established hypoxic microenvironments, as evidenced by the stabilization of hypoxia-inducible factor 1α (HIF1α) and increased blood vessel formation in vitro and in vivo. Furthermore, implanted Co-PLG scaffolds were able to recruit 4T1 metastatic breast cancer cells. These results demonstrate that Co-PLG scaffolds can establish an in vivo hypoxic metastatic niche, providing a novel platform to investigate hypoxic regulation of disseminated tumor cells (DTCs) at target organs.
Collapse
|
67
|
Modulation of Receptor Tyrosine Kinase Activity through Alternative Splicing of Ligands and Receptors in the VEGF-A/VEGFR Axis. Cells 2019; 8:cells8040288. [PMID: 30925751 PMCID: PMC6523102 DOI: 10.3390/cells8040288] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/14/2022] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) signaling is essential for physiological and pathological angiogenesis. Alternative splicing of the VEGF-A pre-mRNA gives rise to a pro-angiogenic family of isoforms with a differing number of amino acids (VEGF-Axxxa), as well as a family of isoforms with anti-angiogenic properties (VEGF-Axxxb). The biological functions of VEGF-A proteins are mediated by a family of cognate protein tyrosine kinase receptors, known as the VEGF receptors (VEGFRs). VEGF-A binds to both VEGFR-1, largely suggested to function as a decoy receptor, and VEGFR-2, the predominant signaling receptor. Both VEGFR-1 and VEGFR-2 can also be alternatively spliced to generate soluble isoforms (sVEGFR-1/sVEGFR-2). The disruption of the splicing of just one of these genes can result in changes to the entire VEGF-A/VEGFR signaling axis, such as the increase in VEGF-A165a relative to VEGF-A165b resulting in increased VEGFR-2 signaling and aberrant angiogenesis in cancer. Research into this signaling axis has recently focused on manipulating the splicing of these genes as a potential therapeutic avenue in disease. Therefore, further research into understanding the mechanisms by which the splicing of VEGF-A/VEGFR-1/VEGFR-2 is regulated will help in the development of drugs aimed at manipulating splicing or inhibiting specific splice isoforms in a therapeutic manner.
Collapse
|
68
|
Bellafiore M, Battaglia G, Bianco A, Palma A. Expression Pattern of Angiogenic Factors in Healthy Heart in Response to Physical Exercise Intensity. Front Physiol 2019; 10:238. [PMID: 30984008 PMCID: PMC6447665 DOI: 10.3389/fphys.2019.00238] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/22/2019] [Indexed: 12/22/2022] Open
Abstract
Recently, many studies showing the regeneration potential of both cardiac and hematopoietic stem cells in adult heart following injury were definitively retracted by the literature. Therefore, stimulating myocardial angiogenesis becomes to be important for preventing cardiovascular diseases. Regular endurance exercise has been reported to induce capillary growth in healthy and diseased myocardium resulting in cardioprotective phenotype. Previously, we demonstrated a significantly increased capillary proliferation in mouse hearts following 30 and 45 days of endurance training. In the present study, we examined the localization and expression pattern of vascular endothelial growth factor receptors (VEGFR-1/Flt-1 and VEGFR-2/Flk-1), hypoxia-inducible factor-1α (HIF-1α), and inducible nitric oxide synthase (iNOS) in heart neocapillarization in response to a mild, moderate, and high intensity of endurance training. Sixty-three Swiss male mice were divided into four untrained control groups and three groups trained for 15 (T15), 30 (T30), and 45 (T45) days with a gradually increasing intensity on a treadmill. We observed the localization of studied proteins with immunostaining and their expression level with Western blot analyses. We found that VEGFR-2/Flk-1 expression progressively increased in trained groups compared with controls, while VEGFR-1/Flt-1 and HIF-1α were higher in T15 than in controls, T30, and T45 animals. Differently, iNOS levels enhanced after 15 and 30 days of exercise. The localization of these factors was not altered by exercise. The results showed that the expression of VEGFR-1/Flt-1, VEGFR-2/Flk-1, HIF-1α, and iNOS is differently regulated in cardiac angiogenesis according to the exercise intensity. VEGFR-1/Flt-1 and HIF-1α are upregulated by a mild intensity exercise, while VEGFR-2/Flk-1 progressively enhances with increasing workload. Differently, iNOS protein is modulated by a moderate intensity exercise. VEGF pathway appears to be involved in exercise-related angiogenesis in heart and VEGF might act in a paracrine and endocrine manner. Understanding this relationship is important for developing exercise strategies to protect the heart by insults.
Collapse
Affiliation(s)
- Marianna Bellafiore
- Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy
| | - Giuseppe Battaglia
- Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy
| | - Antonino Bianco
- Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy
| | - Antonio Palma
- Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy
| |
Collapse
|
69
|
Schito L. Hypoxia-Dependent Angiogenesis and Lymphangiogenesis in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:71-85. [PMID: 31201717 DOI: 10.1007/978-3-030-12734-3_5] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypoxia (low O2) is a ubiquitous feature of solid cancers, arising as a mismatch between cellular O2 supply and consumption. Hypoxia is associated to metastatic disease and mortality owing to its ability to stimulate the formation of blood (angiogenesis) and lymphatic vessels (lymphangiogenesis), thereby allowing cancer cells to escape the unfavorable tumor microenvironment and disseminate into secondary sites. This review outlines molecular mechanisms by which intratumoral hypoxia regulates the expression of motogenic and mitogenic factors that induce angiogenesis and lymphangiogenesis, whilst discussing their implications for metastatic cancers.
Collapse
Affiliation(s)
- Luana Schito
- Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
70
|
Sasagawa T, Nagamatsu T, Morita K, Mimura N, Iriyama T, Fujii T, Shibuya M. HIF-2α, but not HIF-1α, mediates hypoxia-induced up-regulation of Flt-1 gene expression in placental trophoblasts. Sci Rep 2018; 8:17375. [PMID: 30478339 PMCID: PMC6255857 DOI: 10.1038/s41598-018-35745-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/09/2018] [Indexed: 12/14/2022] Open
Abstract
Placental hypoxia and elevated levels of circulating soluble Fms-like tyrosine kinase-1 (sFlt-1), an anti-angiogenic factor, are closely related to the pathogenesis of preeclampsia. Although sFlt-1 secretion from the placental trophoblasts is increased under hypoxic conditions, the underlying molecular mechanism remains unclear. Previously, an authentic hypoxia response element in the Flt-1 gene promoter was shown to be a potential binding site for hypoxia-inducible factors (HIFs). Here, we investigated the roles of HIF-1α and HIF-2α in Flt-1 gene expression in trophoblast-derived choriocarcinoma cell lines and cytotrophoblasts exposed to hypoxic conditions. In the cell lines, increased expression of sFlt-1 splice variants and nuclear accumulation of HIF-1α and HIF-2α were observed after hypoxic stimulation. A specific small interfering RNA or an inhibitor molecule targeting HIF-2α decreased hypoxia-induced up-regulation of Flt-1 gene expression. Moreover, in cytotrophoblasts, increased sFlt-1 mRNA expression and elevated sFlt-1 production were induced by hypoxic stimulation. Notably, hypoxia-induced elevation of sFlt-1 secretion from the cytotrophoblasts was inhibited by silencing the HIF-2α, but not HIF-1α mRNA. These findings suggest that hypoxia-induced activation of HIF-2α is essential for the increased production of sFlt-1 proteins in trophoblasts. Targeting the HIF-2α may be a novel strategy for the treatment of preeclampsia.
Collapse
Affiliation(s)
- Tadashi Sasagawa
- Institute of Physiology and Medicine, Jobu University, Gunma, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Kazuki Morita
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Nobuko Mimura
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Takayuki Iriyama
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Masabumi Shibuya
- Institute of Physiology and Medicine, Jobu University, Gunma, Japan.
| |
Collapse
|
71
|
The role of placental growth factor (PlGF) and its receptor system in retinal vascular diseases. Prog Retin Eye Res 2018; 69:116-136. [PMID: 30385175 DOI: 10.1016/j.preteyeres.2018.10.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/23/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022]
Abstract
Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family. Upon binding to VEGF- and neuropilin-receptor sub-types, PlGF modulates a range of neural, glial and vascular cell responses that are distinct from VEGF-A. As PlGF expression is selectively associated with pathological angiogenesis and inflammation, its blockade does not affect the healthy vasculature. PlGF actions have been extensively described in tumor biology but more recently there has been accumulating preclinical evidence that indicates that this growth factor could have an important role in retinal diseases. High levels of PlGF have been found in aqueous humor, vitreous and/or retina of patients exhibiting retinopathies, especially those with diabetic retinopathy (DR) and neovascular age-related macular degeneration (nvAMD). Expression of this growth factor seems to correlate closely with many of the key pathogenic features of early and late retinopathy in preclinical models. For example, studies using genetic modification and/or pharmacological treatment to block PlGF in the laser-induced choroidal neovascularization (CNV) model, oxygen-induced retinopathy model, as well as various murine diabetic models, have shown that PlGF deletion or inhibition can reduce neovascularization, retinal leakage, inflammation and gliosis, without affecting vascular development or inducing neuronal degeneration. Moreover, an inhibitory effect of PlGF blockade on retinal scarring in the mouse CNV model has also been recently demonstrated and was found to be unique for PlGF inhibition, as compared to various VEGF inhibition strategies. Together, these preclinical results suggest that anti-PlGF therapy might have advantages over anti-VEGF treatment, and that it may have clinical applications as a standalone treatment or in combination with anti-VEGF. Additional clinical studies are clearly needed to further elucidate the role of PlGF and its potential as a therapeutic target in ocular diseases.
Collapse
|
72
|
Davies KTJ, Bennett NC, Faulkes CG, Rossiter SJ. Limited Evidence for Parallel Molecular Adaptations Associated with the Subterranean Niche in Mammals: A Comparative Study of Three Superorders. Mol Biol Evol 2018; 35:2544-2559. [PMID: 30137400 PMCID: PMC6188548 DOI: 10.1093/molbev/msy161] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Among mammals, several lineages have independently adapted to a subterranean niche and possess similar phenotypic traits for burrowing (e.g., cylindrical bodies, short limbs, and absent pinnae). Previous research on mole-rats has revealed molecular adaptations for coping with reduced oxygen, elevated carbon dioxide, and the absence of light. In contrast, almost nothing is known regarding molecular adaptations in other subterranean lineages (e.g., true moles and golden moles). Therefore, the extent to which the recurrent phenotypic adaptations of divergent subterranean taxa have arisen via parallel routes of molecular evolution remains untested. To address these issues, we analyzed ∼8,000 loci in 15 representative subterranean taxa of four independent transitions to an underground niche for signatures of positive selection and convergent amino acid substitutions. Complementary analyses were performed in nonsubterranean "control" taxa to assess the biological significance of results. We found comparable numbers of positively selected genes in each of the four subterranean groups; however, correspondence in terms of gene identity between gene sets was low. Furthermore, we did not detect evidence of more convergent amino acids among subterranean species pairs compared with levels found between nonsubterranean controls. Comparisons with nonsubterranean taxa also revealed loci either under positive selection or with convergent substitutions, with similar functional enrichment (e.g., cell adhesion, immune response, and coagulation). Given the limited indication that positive selection and convergence occurred in the same loci, we conclude that selection may have acted on different loci across subterranean mammal lineages to produce similar phenotypes.
Collapse
Affiliation(s)
- Kalina T J Davies
- School of Biological & Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Nigel C Bennett
- Department of Zoology & Entomology, Mammal Research Institute, University of Pretoria, Pretoria, South Africa
| | - Chris G Faulkes
- School of Biological & Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Stephen J Rossiter
- School of Biological & Chemical Sciences, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
73
|
Banadakoppa M, Balakrishnan M, Yallampalli C. Upregulation and release of soluble fms-like tyrosine kinase receptor 1 mediated by complement activation in human syncytiotrophoblast cells. Am J Reprod Immunol 2018; 80:e13033. [PMID: 30099798 DOI: 10.1111/aji.13033] [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: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 01/01/2023] Open
Abstract
PROBLEM Antiangiogenic molecule soluble fms-like tyrosine kinase receptor 1 (sFLT1) released from trophoblast cells is associated with pregnancy-specific hypertensive disorder pre-eclampsia. Cause of elevated sFLT1 in pre-eclampsia patients is not well understood. Despite evidence of excess systemic and placental complement activation in pre-eclampsia patients, its role in pathophysiology is not clear. If the complement activation plays a role in upregulation and secretion of sFLT1 is not known. METHOD OF STUDY Human trophoblast cells were isolated from term placentas and allowed to syncytialize. Complement was activated in vitro at sublethal levels on syncytiotrophoblast cells. Effect of complement activation on expression and release of sFLT1 was assessed by comparing its levels in these cells with and without complement activation. RESULTS Sublethal level of complement activation on syncytialized human trophoblast cells induced upregulation of sFLT1 mRNA and protein. Complement also induced secretion of sFLT1 in a manner depending on degree of activation. Anaphylatoxins C3a induced upregulation but not the release of sFLT1. Release of terminal membrane attack complex (MAC) was associated with sFLT1 secretion. CONCLUSION Complement activation plays a major role in both the expression and secretion of sFLT1 from syncytial trophoblast cells. The terminal MAC complex is involved in its secretion. Increased levels of sFLT1 in pre-eclampsia patients may be due to complement-induced upregulation and secretion.
Collapse
Affiliation(s)
- Manu Banadakoppa
- Basic Sciences Perinatology Research Laboratories, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas
| | - Meena Balakrishnan
- Basic Sciences Perinatology Research Laboratories, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas
| | - Chandra Yallampalli
- Basic Sciences Perinatology Research Laboratories, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
74
|
Kotb MA, Bedewi MA, Aldossary NM, Mahmoud G, Naguib MF. Sonographic assessment of carpal tunnel syndrome in diabetic patients with and without polyneuropathy. Medicine (Baltimore) 2018; 97:e11104. [PMID: 29901629 PMCID: PMC6024071 DOI: 10.1097/md.0000000000011104] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The objective of this study is to determine whether the cross sectional area (CSA) measurement of the median nerve at the wrist differ between carpal tunnel syndrome (CTS) in diabetic patients with and without diabetic polyneuropathy (DPN).This study included 44 patients with type II diabete millitus (DM) with CTS, 32 patients with CTS and DPN, 46 patients with idiopathic CTS, and 42 healthy subjects. Ultrasonographic measurement of the CSA of the median nerve was made at the level of the wrist, together with nerve conduction studies.The median CSA at the wrist was significantly larger in all patient groups compared with healthy subjects. The median nerve CSA was significantly larger in diabetic patients with CTS than patients with idiopathic CTS. The median nerve CSA at wrist was significantly smaller in patients with CTS and DPN compared with diabetic patients with CTS only.The median nerve CSA at the wrist was larger in diabetic patients with CTS than patients with idiopathic CTS and CTS with DPN. Median nerve CSA can help to differentiate between diabetic patients with CTS with and without DPN.
Collapse
Affiliation(s)
- Mamdouh Ali Kotb
- College of Medicine, Prince Sattam bin Abdulaziz University, Al-kharj, Kingdom of Saudi Arabia
- Neurology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | | | - Nasser M. Aldossary
- College of Medicine, Prince Sattam bin Abdulaziz University, Al-kharj, Kingdom of Saudi Arabia
| | - Gehan Mahmoud
- College of Medicine, Prince Sattam bin Abdulaziz University, Al-kharj, Kingdom of Saudi Arabia
| | | |
Collapse
|
75
|
Liu J, Hou W, Guan T, Tang L, Zhu X, Li Y, Hou S, Zhang J, Chen H, Huang Y. Slit2/Robo1 signaling is involved in angiogenesis of glomerular endothelial cells exposed to a diabetic-like environment. Angiogenesis 2018; 21:237-249. [PMID: 29299781 DOI: 10.1007/s10456-017-9592-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 12/26/2017] [Indexed: 01/08/2023]
Abstract
Abnormal angiogenesis plays a pathological role in diabetic nephropathy (DN), contributing to glomerular hypertrophy and microalbuminuria. Slit2/Robo1 signaling participates in angiogenesis in some pathological contexts, but whether it is involved in glomerular abnormal angiogenesis of early DN is unclear. The present study evaluated the effects of Slit2/Robo1 signaling pathway on angiogenesis of human renal glomerular endothelial cells (HRGECs) exposed to a diabetic-like environment or recombinant Slit2-N. To remove the effect of Slit2 derived from mesangial cells, human renal mesangial cells (HRMCs) grown in high glucose (HG) medium (33 mM) were transfected with Slit2 siRNA and then the HG-HRMCs-CM with Slit2 depletion was collected after 48 h. HRGECs were cultured in the HG-HRMCs-CM or recombinant Slit2-N for 0, 6, 12, 24, or 48 h. The mRNA and protein expressions of Slit2/Robo1, PI3K/Akt and HIF-1α/VEGF signaling pathways were detected by quantitative real-time PCR, western blotting, and ELISA, respectively. The CCK-8 cell proliferation assay, flow cytometry and the scratch wound-healing assay were used to assess cell proliferation, cycles, and migration, respectively. Matrigel was used to perform a tubule formation assay. Our results showed that the HG-HRMCs-CM with Slit2 depletion enhanced the activation of Slit2/Robo1, PI3K/Akt, and HIF-1α/VEGF signaling in HRGECs in time-dependent manner (0-24 h post-treatment). In addition, the HG-HRMCs-CM with Slit2 depletion significantly promoted HRGECs proliferation, migration, and tube formation. Pretreatment of HRGECs with Robo1 siRNA suppressed the activation of PI3K/Akt and HIF-1α/VEGF signaling and inhibited angiogenesis, whereas PI3K inhibitor suppressed HIF-1α/VEGF signaling, without influencing Robo1 expression. In the HRGECs treated with Slit2-N, Slit2-N time-dependently enhanced the activation of Robo1/PI3K/Akt/VEGF pathway but not HIF-1α activity, and promoted HRGECs proliferation, migration, and tube formation. The effects induced by Slit2 were also abolished by Robo1 siRNA and PI3K inhibitor. Taken together, our findings indicate that in a diabetic-like environment, in addition to mesangial cells, autocrine activation of Slit2/Robo1 signaling of HRGECs may contribute to angiogenesis of HRGECs through PI3K/Akt/VEGF pathway; therefore, Slit2/Robo1 signaling may be a potent therapeutic target for the treatment of abnormal angiogenesis in early DN and may have broad implications for the treatment of other diseases dependent on pathologic angiogenesis.
Collapse
Affiliation(s)
- Junhui Liu
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Weiping Hou
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Tao Guan
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Luyao Tang
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xufei Zhu
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yi Li
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Shihui Hou
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Jun Zhang
- Department of Pathology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Hua Chen
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yunjian Huang
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
76
|
Failla CM, Carbo M, Morea V. Positive and Negative Regulation of Angiogenesis by Soluble Vascular Endothelial Growth Factor Receptor-1. Int J Mol Sci 2018; 19:ijms19051306. [PMID: 29702562 PMCID: PMC5983705 DOI: 10.3390/ijms19051306] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 12/20/2022] Open
Abstract
Vascular endothelial growth factor receptor (VEGFR)-1 exists in different forms, derived from alternative splicing of the same gene. In addition to the transmembrane form, endothelial cells produce a soluble VEGFR-1 (sVEGFR-1) isoform, whereas non-endothelial cells produce both sVEGFR-1 and a different soluble molecule, known as soluble fms-like tyrosine kinase (sFlt)1-14. By binding members of the vascular endothelial growth factor (VEGF) family, the soluble forms reduce the amounts of VEGFs available for the interaction with their transmembrane receptors, thereby negatively regulating VEGFR-mediated signaling. In agreement with this activity, high levels of circulating sVEGFR-1 or sFlt1-14 are associated with different pathological conditions involving vascular dysfunction. Moreover, sVEGFR-1 and sFlt1-14 have an additional role in angiogenesis: they are deposited in the endothelial cell and pericyte extracellular matrix, and interact with cell membrane components. Interaction of sVEGFR-1 with α5β1 integrin on endothelial cell membranes regulates vessel growth, triggering a dynamic, pro-angiogenic phenotype. Interaction of sVEGFR-1/sFlt1-14 with cell membrane glycosphingolipids in lipid rafts controls kidney cell morphology and glomerular barrier functions. These cell⁻matrix contacts represent attractive novel targets for pharmacological intervention in addition to those addressing interactions between VEGFs and their receptors.
Collapse
Affiliation(s)
| | - Miriam Carbo
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University, 00185 Rome, Italy.
| | - Veronica Morea
- National Research Council of Italy (CNR), Department of Biochemical Sciences "A. Rossi Fanelli", Institute of Molecular Biology and Pathology c/o, Sapienza University, 00185 Rome, Italy.
| |
Collapse
|
77
|
Pan X, Gong D, Gao F, Sangild PT. Diet-dependent changes in the intestinal DNA methylome after introduction of enteral feeding in preterm pigs. Epigenomics 2018; 10:395-408. [PMID: 29587528 DOI: 10.2217/epi-2017-0122] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM To examine how enteral feeding affects the intestinal epigenome and gene expression just after preterm birth. MATERIALS & METHODS Intestinal tissue from preterm pigs, modeling preterm infants, was collected at birth and 5 days after gradual introduction of infant formula or bovine colostrum. The intestinal tissue was analyzed by reduced representation bisulfite sequencing and real-time qPCR. RESULTS Relative to colostrum, formula increased bacterial epithelial adherence and lipopolysaccharide binding protein (LBP) expression, which was regulated by promoter methylation. Diet-dependent changes in DNA methylation and/or mRNA expression were related to innate immune response, hypoxia, angiogenesis and epithelial-mesenchymal transition pathways (e.g., TTC38, IL8, C3, HIF1A and VEGFR1). CONCLUSION Epigenetic changes may mediate important effects of the first feeding on intestinal development in preterm neonates.
Collapse
Affiliation(s)
- Xiaoyu Pan
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg DK 1870 C, Denmark
| | - Desheng Gong
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, PR China
| | - Fei Gao
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, PR China
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg DK 1870 C, Denmark
| |
Collapse
|
78
|
Tudisco L, Orlandi A, Tarallo V, De Falco S. Hypoxia activates placental growth factor expression in lymphatic endothelial cells. Oncotarget 2018; 8:32873-32883. [PMID: 28427198 PMCID: PMC5464835 DOI: 10.18632/oncotarget.15861] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/08/2017] [Indexed: 01/13/2023] Open
Abstract
Placental growth factor (PlGF), a proangiogenic member of vascular endothelial growth family, is active during pathological conditions like cancer, metastasis formation and hind limb ischemia and in wound healing. Endothelial cells express PlGF and hypoxia positively modulates in vitro its expression. To verify whether hypoxia modulates PlGF expression in different cellular contexts and in vivo, we first analyzed five human and five mouse cancer cell lines showing that in eight of them hypoxia positively modulates PlGF. Next, we analyzed xenograft colorectal cancer tumors showing that human cancer cells were able to express PlGF in hypoxic area of the tumor. Surprisingly, we did not visualize mouse PlGF in CD31 positive tumor vessels, but in low CD31 positive vessels, a characteristic of lymphatic vessels. We found that hypoxia effectively activates PlGF expression in lymphatic endothelial cells as well as in LYVE1 positive tumor vessels. We also investigated two additional mouse angiogenic models, hind limb ischemia and wound healing, and we confirmed that lymphatic vessels of both ischemic muscles and skin express PlGF. These results show for the first time that hypoxia activates PlGF expression in lymphatic endothelial cells, which have to be considered an additional source for PlGF production in pathological contexts.
Collapse
Affiliation(s)
- Laura Tudisco
- Angiogenesis LAB, Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso', CNR, Naples, Italy
| | - Augusto Orlandi
- Department of Biomedicine and Prevention, Anatomic Pathology, University of Tor Vergata, Rome, Italy
| | - Valeria Tarallo
- Angiogenesis LAB, Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso', CNR, Naples, Italy
| | - Sandro De Falco
- Angiogenesis LAB, Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso', CNR, Naples, Italy
| |
Collapse
|
79
|
Adebambo OA, Shea D, Fry RC. Cadmium disrupts signaling of the hypoxia-inducible (HIF) and transforming growth factor (TGF-β) pathways in placental JEG-3 trophoblast cells via reactive oxygen species. Toxicol Appl Pharmacol 2018; 342:108-115. [DOI: 10.1016/j.taap.2018.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/09/2018] [Accepted: 01/17/2018] [Indexed: 12/16/2022]
|
80
|
Palmer KR, Tong S, Kaitu'u-Lino TJ. Placental-specific sFLT-1: role in pre-eclamptic pathophysiology and its translational possibilities for clinical prediction and diagnosis. Mol Hum Reprod 2018; 23:69-78. [PMID: 27986932 DOI: 10.1093/molehr/gaw077] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/09/2016] [Indexed: 11/12/2022] Open
Abstract
Pre-eclampsia is a common obstetric complication globally responsible for a significant burden of maternal and perinatal morbidity and mortality. Central to its pathophysiology is the anti-angiogenic protein, soluble fms-like tyrosine kinase-1 (sFLT-1). sFLT-1 is released from a range of tissues into the circulation, where it antagonizes the activity of vascular endothelial growth factor and placental growth factor leading to endothelial dysfunction. It is this widespread endothelial dysfunction that produces the clinical features of pre-eclampsia including hypertension and proteinuria. There are multiple splice variants of sFLT-1. One, known as sFLT-1 e15a, evolved quite recently and is only present in humans and higher order primates. This sFLT-1 variant is also the main sFLT-1 secreted from the placenta. Recent work has shown that sFLT-1 e15a is significantly elevated in the placenta and circulation of women with pre-eclampsia. It is also biologically active, capable of causing endothelial dysfunction and the end-organ dysfunction seen in pre-eclampsia. Indeed, the over-expression of sFLT-1 e15a in mice recapitulates the pre-eclamptic phenotype in pregnancy. Therefore, here we propose that sFLT-1 e15a may be the sFLT-1 variant primarily responsible for pre-eclampsia, a uniquely human disease. Furthermore, this placental-specific sFLT-1 variant provides promise for use as an accurate biomarker in the prediction or diagnosis of pre-eclampsia.
Collapse
Affiliation(s)
- K R Palmer
- Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, 246 Clayton Rd, Clayton, 3168 Victoria, Australia.,Translational Obstetric Group, University of Melbourne, Mercy Hospital for Women, 163 Studley Rd, Heidelberg, 3084 Victoria, Australia
| | - S Tong
- Translational Obstetric Group, University of Melbourne, Mercy Hospital for Women, 163 Studley Rd, Heidelberg, 3084 Victoria, Australia
| | - T J Kaitu'u-Lino
- Translational Obstetric Group, University of Melbourne, Mercy Hospital for Women, 163 Studley Rd, Heidelberg, 3084 Victoria, Australia
| |
Collapse
|
81
|
Genome-wide profiling of gene expression and DNA methylation provides insight into low-altitude acclimation in Tibetan pigs. Gene 2018; 642:522-532. [DOI: 10.1016/j.gene.2017.11.074] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 11/24/2017] [Accepted: 11/28/2017] [Indexed: 02/06/2023]
|
82
|
Ugurlu T, Ozogul C, Saribas GS, Gurgen SG, Akyol SN, Kartal B. The effect of antioxidants on angiogenesis in uterine transplantation. J OBSTET GYNAECOL 2018; 38:382-387. [DOI: 10.1080/01443615.2017.1316250] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Tugba Ugurlu
- Faculty of Medicine, Department of Histology and Embryology, Gazi University, Ankara, Turkey
| | - Candan Ozogul
- Faculty of Medicine, Department of Histology and Embryology, Gazi University, Ankara, Turkey
| | - Gulistan Sanem Saribas
- Faculty of Medicine, Department of Histology and Embryology, Gazi University, Ankara, Turkey
| | - Seren Gulsen Gurgen
- Department of Histology and Embryology, School of Vocational Health Services, Celal Bayar University, Manisa, Turkey
| | - Seda Nur Akyol
- Faculty of Medicine, Department of Histology and Embryology, Gazi University, Ankara, Turkey
| | - Bahar Kartal
- Faculty of Medicine, Department of Histology and Embryology, Gazi University, Ankara, Turkey
- Department of Histology and Embryology, Kafkas University Medical Faculty, Kars, Turkey
| |
Collapse
|
83
|
Barratt SL, Blythe T, Ourradi K, Jarrett C, Welsh GI, Bates DO, Millar AB. Effects of hypoxia and hyperoxia on the differential expression of VEGF-A isoforms and receptors in Idiopathic Pulmonary Fibrosis (IPF). Respir Res 2018; 19:9. [PMID: 29334947 PMCID: PMC5769544 DOI: 10.1186/s12931-017-0711-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 12/26/2017] [Indexed: 12/20/2022] Open
Abstract
Dysregulation of VEGF-A bioavailability has been implicated in the development of lung injury/fibrosis, exemplified by Idiopathic Pulmonary Fibrosis (IPF). VEGF-A is a target of the hypoxic response via its translational regulation by HIF-1α. The role of hypoxia and hyperoxia in the development and progression of IPF has not been explored. In normal lung (NF) and IPF-derived fibroblasts (FF) VEGF-Axxxa protein expression was upregulated by hypoxia, mediated through activation of VEGF-Axxxa gene transcription. VEGF-A receptors and co-receptors were differentially expressed by hypoxia and hyperoxia. Our data supports a potential role for hypoxia, hyperoxia and VEGF-Axxxa isoforms as drivers of fibrogenesis.
Collapse
Affiliation(s)
- Shaney L Barratt
- Academic Respiratory Unit, Learning and Research Building, Southmead Hospital, Bristol, UK.
| | - Thomas Blythe
- Academic Respiratory Unit, Learning and Research Building, Southmead Hospital, Bristol, UK
| | - Khadija Ourradi
- Academic Respiratory Unit, Learning and Research Building, Southmead Hospital, Bristol, UK
| | - Caroline Jarrett
- Academic Respiratory Unit, Learning and Research Building, Southmead Hospital, Bristol, UK
| | - Gavin I Welsh
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - David O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ann B Millar
- Academic Respiratory Unit, Learning and Research Building, Southmead Hospital, Bristol, UK
| |
Collapse
|
84
|
Hoar FJ, Lip GYH, Belgore F, Stonelake PS. Circulating Levels of Vegf-A, VEGF-D and soluble VEGF-A Receptor (sFlt-1) in Human Breast Cancer. Int J Biol Markers 2018; 19:229-35. [PMID: 15503825 DOI: 10.1177/172460080401900308] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
As circulating levels of vascular endothelial growth factor (VEGF-A) are raised in malignancy, the aim of this study was to investigate whether similar changes occur in two related factors, VEGF-D and the soluble VEGF-A receptor Flt-1 (sFlt-1). Circulating levels of VEGF-A, VEGF-D and sFlt-1 were determined by ELISA in 51 patients with primary breast cancer and matched healthy controls. Results were correlated with clinicopathological data. Whilst there was a difference in VEGF-A levels between patient and control groups (p=0.03), no such difference was observed for sFlt-1 or VEGF-D levels and there was no association between individual factors and the clinicopathological variables examined. However, there was a positive correlation between VEGF-A and sFlt-1 levels in both patient and control groups (p<0.0001). In addition, the ratio of sFlt-1 to VEGF-A was significantly different between patients and controls (p<0.0001) and was also associated with tumour size (p=0.01) within the patient group. During tumour progression there is a change in the relative amounts of sFlt-1 and VEGF-A in the circulation. Measuring the sFlt-1:VEGF-A ratio may have more significance than VEGF-A alone and further studies are needed to determine whether the ratio is of use as a prognostic marker or as a means of monitoring response to anti-angiogenic therapy in cancer.
Collapse
Affiliation(s)
- F J Hoar
- Haemostasis Thrombosis and Vascular Biology Unit, University Department of Medicine, City Hospital, Birmingham--U.K
| | | | | | | |
Collapse
|
85
|
Abstract
Vascular endothelial growth factor (VEGF) is a potent angiogenic factor. Despite upregulation of VEGF in the brain in Alzheimer's disease (AD), probably in response to amyloid-β, vasoconstriction, and tissue hypoxia, there is no consequent increase in microvessel density. VEGF binds to and activates VEGF receptor 2 (VEGFR2), but also binds to VEGF receptor 1 (VEGFR1), which exists in less-active membrane-bound and inactive soluble (sVEGFR1) forms and inhibits pro-angiogenic signaling. We have investigated whether altered expression of VEGF receptors might account for the lack of angiogenic response to VEGF in AD. We assessed the cellular distribution and protein level of VEGFR1 and VEGFR2 in parietal cortex from 50 AD and 36 age-matched control brains, and related the findings to measurements of VEGF and von Willebrand factor level (a marker of microvessel density) in the same tissue samples. VEGFR2 was expressed by neurons, astrocytes and endothelial cells. VEGFR1 was expressed predominantly neuronally and was significantly reduced in AD (p = 0.02). Western blot analysis on a subset of brains showed reduction in VEGFR1:sVEGFR1 in AD (p = 0.046). The lack of angiogenesis despite cerebral hypoperfusion in AD is not explained by altered expression of VEGFR2 or total VEGFR1; indeed, the downregulation of VEGFR1 may represent a pro-angiogenic response to the hypoperfusion. However, the relative increase in sVEGFR1 would be expected to have an anti-angiogenic effect which may be a factor in AD.
Collapse
Affiliation(s)
- Rachel Harris
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| | - James Scott Miners
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| | - Shelley Allen
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| | - Seth Love
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| |
Collapse
|
86
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
87
|
Characterization of neural stem cells modified with hypoxia/neuron-specific VEGF expression system for spinal cord injury. Gene Ther 2017; 25:27-38. [PMID: 29155421 DOI: 10.1038/gt.2017.92] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 09/30/2017] [Accepted: 10/04/2017] [Indexed: 02/01/2023]
Abstract
Spinal cord injury (SCI) is an incurable disease causing an ischemic environment and functional defect, thus a new therapeutic approach is needed for SCI treatment. Vascular endothelial growth factor (VEGF) is a potent therapeutic gene to treat SCI via angiogenesis and neuroprotection, and both tissue-specific gene expression and high gene delivery efficiency are important for successful gene therapy. Here we design the hypoxia/neuron dual-specific gene expression system (pEpo-NSE) and efficient gene delivery platform can be achieved by the combination ex vivo gene therapy with erythropoietin (Epo) enhancer, neuron-specific enolase (NSE) promoter and neural stem cells (NSCs). An in vitro model, NSCs transfected with pEpo-NSE were consistently and selectively overexpressing therapeutic genes in response to neural differentiation and hypoxic conditions. Also, in SCI model, ex vivo gene therapy using pEpo-NSE system with NSCs significantly enhanced gene delivery efficiency compared with pEpo-NSE system gene therapy alone. However, microarray analysis reveals that introducing exogenous pEpo-NSE and VEGF triggers biological pathways in NSCs such as glycolysis and signaling pathways such as Ras and mitogen-activated protein kinase, leading to cell proliferation, differentiation and apoptosis. Collectively, it indicates that the pEpo-NSE gene expression system works stably in NSCs and ex vivo gene therapy using pEpo-NSE system with NSCs improves gene expression efficiency. However, exogenously introduced pEpo-NSE system has an influence on gene expression profiles in NSCs. Therefore, when we consider ex vivo gene therapy for SCI, the effects of changes in gene expression profiles in NSCs on safety should be investigated.
Collapse
|
88
|
Zhang W, Zhao X, Xiao Y, Chen J, Han P, Zhang J, Fu H, James Kang Y. The association of depressed angiogenic factors with reduced capillary density in the Rhesus monkey model of myocardial ischemia. Metallomics 2017; 8:654-62. [PMID: 26852735 DOI: 10.1039/c5mt00332f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Depressed capillary density is associated with myocardial ischemic infarction, in which hypoxia-inducible factor 1α (HIF-1α) is increased. The present study was undertaken to examine changes in the angiogenic factors whose expression is regulated by HIF-1 and their relation to the depressed capillary density in the Rhesus monkey model of myocardial ischemic infarction. Male Rhesus monkeys 2-3 years old were subjected to myocardial ischemia by permanent ligation of left anterior descending (LAD) artery leading to the development of myocardial infarction. Eight weeks after LAD ligation, copper concentrations, myocardial histological changes and capillary density were examined, along with Western blot and immunohistochemical analysis of angiogenic factors and detection of HIF-1 activity. Capillary density was significantly decreased but the concentrations of HIF-1α and HIF-1β were significantly increased in the infarct area. However, the levels of mRNA and protein for VEGF and VEGFR1 were significantly decreased. Other HIF-1 regulated angiogenic factors, including Tie-2, Ang-1 and FGF-1, were also significantly depressed, but vascular destabilizing factor Ang-2 was significantly increased. Copper concentrations were depressed in the infarct area. Copper-independent HIF-1 activity was increased shown by the elevated mRNA level of IGF-2, a HIF-1 target gene. Removal of copper by a copper chelator, tetraethylenepentamine, from primary cultures of neonatal rat cardiomyocytes also suppressed the expression of HIF-1 regulated VEGF and BNIP3, but not IGF-2. The data suggest that under ischemic conditions, copper loss suppressed the expression of critical angiogenic genes regulated by HIF-1, but did not affect copper-independent HIF-1 activation of gene expression. This copper-dependent dysregulation of angiogenic gene expression would contribute to the pathogenesis of myocardial ischemic infarction.
Collapse
Affiliation(s)
- Wenjing Zhang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Xinmei Zhao
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Ying Xiao
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Jianmin Chen
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Pengfei Han
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Jingyao Zhang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Haiying Fu
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Y James Kang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China. and Department of Pharmacology and Toxicology, University of Louisville, School of Medicine, Louisville, Kentucky 40202, USA
| |
Collapse
|
89
|
Mechanisms of macular edema: Beyond the surface. Prog Retin Eye Res 2017; 63:20-68. [PMID: 29126927 DOI: 10.1016/j.preteyeres.2017.10.006] [Citation(s) in RCA: 413] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/24/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023]
Abstract
Macular edema consists of intra- or subretinal fluid accumulation in the macular region. It occurs during the course of numerous retinal disorders and can cause severe impairment of central vision. Major causes of macular edema include diabetes, branch and central retinal vein occlusion, choroidal neovascularization, posterior uveitis, postoperative inflammation and central serous chorioretinopathy. The healthy retina is maintained in a relatively dehydrated, transparent state compatible with optimal light transmission by multiple active and passive systems. Fluid accumulation results from an imbalance between processes governing fluid entry and exit, and is driven by Starling equation when inner or outer blood-retinal barriers are disrupted. The multiple and intricate mechanisms involved in retinal hydro-ionic homeostasis, their molecular and cellular basis, and how their deregulation lead to retinal edema, are addressed in this review. Analyzing the distribution of junction proteins and water channels in the human macula, several hypotheses are raised to explain why edema forms specifically in the macular region. "Pure" clinical phenotypes of macular edema, that result presumably from a single causative mechanism, are detailed. Finally, diabetic macular edema is investigated, as a complex multifactorial pathogenic example. This comprehensive review on the current understanding of macular edema and its mechanisms opens perspectives to identify new preventive and therapeutic strategies for this sight-threatening condition.
Collapse
|
90
|
Chen ZY, Yao Y. A synergistic negative effect of gestational smoke-exposure and small litter size on rat placental efficiency, vascularisation and angiogenic factors mRNA expression. PLoS One 2017; 12:e0181348. [PMID: 28719646 PMCID: PMC5515421 DOI: 10.1371/journal.pone.0181348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/14/2017] [Indexed: 01/09/2023] Open
Abstract
Smoking increases the risk of pregnancy complications such as spontaneous abortion and low birth weight (LBW). By cigarette smoke exposure (gestational day, GD3-17), normal-litter-size pregnancy with low birth weight (NP-LBW) and small-litter-size pregnancy with normal birth weight (SP-NBW) in rats were induced. The placental weight in SP-NBW was twice the weight of the normal in contrast with the smaller placenta in NP-LBW. Compared with the normal, placental efficiency (expressed as fetus-to-placenta weight ratio) and placental vascularisation were significantly decreased in smoke exposed placentas with more obvious decrease in SP-NBW. For NP-LBW, decreased placental vascularisation was due to decreased labyrinth vascularisation which was caused by both decreased number density and diameter of fetal capillary. For SP-NBW, decreased placental vascularisation was due to reduced proportion of labyrinth in placenta and decreased labyrinth vascularisation which was caused by decreased fetal capillary number density. Real time RT-PCR analysis showed a tendency for decreased placental mRNA level of vascular endothelial growth factor (VEGF), angiopoietin-1 (Ang1) and tyrosine kinase receptor-2 (Tie2) in NP-LBW(P<0.1), and the tendency became obvious in SP-NBW(P<0.05). A tendency for decreased placental mRNA level of fms-like tyrosine kinase-1(Flt1) and angiopoietin-2 (Ang2) was also observed in SP-LBW(P<0.1). Our data demonstrated the synergistic negative effect of gestational smoke-exposure and small litter size on placental efficiency, placental vascularisation and placental angiogenic growth factor mRNA expression in rat.
Collapse
Affiliation(s)
- Zhen-Yan Chen
- Department of Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan, Hubei, China
| | - Ying Yao
- Department of Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
91
|
Abstract
In this review we note that the placenta and cancer both develop in microenvironments in which there are gradients of oxygen availability. Whilst fundamentally different in that placental development is organised and physiological whilst cancer is chaotic and pathological, there are similarities in their respective capacities to proliferate, invade adjacent tissues, generate a blood supply and avoid rejection by the immune system. We provide a brief description of the hypoxia-inducible factor (HIF) pathway and indicate the ways by which HIF activity can be regulated to achieve oxygen homeostasis. We then exemplify the potential role of the HIF pathway in contributing to those functions shared between the placenta and cancer through effects on cellular proliferation, cell death, angiogenesis, blood vessel co-option, vascular mimicry, cell adhesion molecules, secretion of matrix metalloproteinases, antigen presentation mechanisms and immunosuppressive factors. We advocate future studies to explore these similarities and differences in the hope of improving our understanding of both systems and hence treatments of placental disorders and cancer.
Collapse
|
92
|
Shiratsuki S, Hara T, Munakata Y, Shirasuna K, Kuwayama T, Iwata H. Low oxygen level increases proliferation and metabolic changes in bovine granulosa cells. Mol Cell Endocrinol 2016; 437:75-85. [PMID: 27519633 DOI: 10.1016/j.mce.2016.08.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/06/2016] [Accepted: 08/08/2016] [Indexed: 01/04/2023]
Abstract
The present study addresses molecular backgrounds underlying low oxygen induced metabolic changes and 1.2-fold change in bovine granulosa cell (GCs) proliferation. RNA-seq revealed that low oxygen (5%) upregulated genes associated with HIF-1 and glycolysis and downregulated genes associated with mitochondrial respiration than that in high oxygen level (21%). Low oxygen level induced high glycolytic activity and low mitochondrial function and biogenesis. Low oxygen level enhanced GC proliferation with high expression levels of HIF-1, VEGF, AKT, mTOR, and S6RP, whereas addition of anti-VEGF antibody decreased cellular proliferation with low phosphorylated AKT and mTOR expression levels. Low oxygen level reduced SIRT1, whereas activation of SIRT1 by resveratrol increased mitochondrial replication and decreased cellular proliferation with reduction of phosphorylated mTOR. These results suggest that low oxygen level stimulates the HIF1-VEGF-AKT-mTOR pathway and up-regulates glycolysis, which contributes to GC proliferation, and downregulation of SIRT1 contributes to hypoxia-associated reduction of mitochondria and cellular proliferation.
Collapse
Affiliation(s)
- Shogo Shiratsuki
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan
| | - Tomotaka Hara
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan
| | - Yasuhisa Munakata
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan
| | - Koumei Shirasuna
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan
| | - Takehito Kuwayama
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan
| | - Hisataka Iwata
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan.
| |
Collapse
|
93
|
Krishnan S, Szabo E, Burghardt I, Frei K, Tabatabai G, Weller M. Modulation of cerebral endothelial cell function by TGF-β in glioblastoma: VEGF-dependent angiogenesis versus endothelial mesenchymal transition. Oncotarget 2016; 6:22480-95. [PMID: 26090865 PMCID: PMC4673177 DOI: 10.18632/oncotarget.4310] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/03/2015] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma are among the most angiogenic tumors. The molecular mechanisms that control blood vessel formation by endothelial cells (EC) in glioblastoma remain incompletely understood. Transforming growth factor-β (TGF-β) is a key regulatory cytokine that has proinvasive and stemness-maintaining autocrine properties in glioblastoma and confers immunosuppression to the tumor microenvironment. Here we characterize potential pro- and anti-angiogenic activities of TGF-β in the context of glioblastoma in vitro, using human brain-derived microvascular endothelial cells (hCMEC/D3) and glioblastoma-derived endothelial cells (GMEC) as model systems. We find that TGF-β induces vascular endothelial growth factor (VEGF) and placental growth factor (PlGF) mRNA expression and protein release in a TGF-β receptor (TβR) II / activin-like kinase (ALK)-5-dependent manner under normoxia and hypoxia, defining potential indirect proangiogenic activity of TGF-β in glioblastoma. In parallel, exogenous TGF-β has also inhibitory effects on EC properties and induces endothelial-mesenchymal transition (EndMT) in hCMEC and GMEC. Accordingly, direct inhibition of endogenous TGF-β/ALK-5 signalling increases EC properties such as tube formation, von-Willebrand factor (vWF) and claudin (CLDN) 5 expression. Yet, the supernatant of TGF-β-stimulated hCMEC and GMEC strongly promotes EC-related gene expression and tube formation in a cediranib-sensitive manner. These observations shed light on the complex pro- and anti-angiogenic pathways involving the cross-talk between TGF-β and VEGF/PLGF signalling in glioblastoma which may involve parallel stimulation of angiogenesis and EndMT in distinct target cell populations.
Collapse
Affiliation(s)
- Shanmugarajan Krishnan
- Laboratory of Molecular Neuro-Oncology, Department of Neurology and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Emese Szabo
- Laboratory of Molecular Neuro-Oncology, Department of Neurology and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Isabel Burghardt
- Laboratory of Molecular Neuro-Oncology, Department of Neurology and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Karl Frei
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Ghazaleh Tabatabai
- Laboratory of Molecular Neuro-Oncology, Department of Neurology and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.,Interdisciplinary Division of Neuro-Oncology, Departments of Vascular Neurology and Neurosurgery, University Hospital Tübingen, Tübingen, Germany
| | - Michael Weller
- Laboratory of Molecular Neuro-Oncology, Department of Neurology and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
94
|
Kraft BD, Suliman HB, Colman EC, Mahmood K, Hartwig MG, Piantadosi CA, Shofer SL. Hypoxic Gene Expression of Donor Bronchi Linked to Airway Complications after Lung Transplantation. Am J Respir Crit Care Med 2016; 193:552-60. [PMID: 26488115 DOI: 10.1164/rccm.201508-1634oc] [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] [Indexed: 12/26/2022] Open
Abstract
RATIONALE Central airway stenosis (CAS) after lung transplantation has been attributed in part to chronic airway ischemia; however, little is known about the time course or significance of large airway hypoxia early after transplantation. OBJECTIVES To evaluate large airway oxygenation and hypoxic gene expression during the first month after lung transplantation and their relation to airway complications. METHODS Subjects who underwent lung transplantation underwent endobronchial tissue oximetry of native and donor bronchi at 0, 3, and 30 days after transplantation (n = 11) and/or endobronchial biopsies (n = 14) at 30 days for real-time polymerase chain reaction of hypoxia-inducible genes. Patients were monitored for 6 months for the development of transplant-related complications. MEASUREMENTS AND MAIN RESULTS Compared with native endobronchial tissues, donor tissue oxygen saturations (Sto2) were reduced in the upper lobes (74.1 ± 1.8% vs. 68.8 ± 1.7%; P < 0.05) and lower lobes (75.6 ± 1.6% vs. 71.5 ± 1.8%; P = 0.065) at 30 days post-transplantation. Donor upper lobe and subcarina Sto2 levels were also lower than the main carina (difference of -3.9 ± 1.5 and -4.8 ± 2.1, respectively; P < 0.05) at 30 days. Up-regulation of hypoxia-inducible genes VEGFA, FLT1, VEGFC, HMOX1, and TIE2 was significant in donor airways relative to native airways (all P < 0.05). VEGFA, KDR, and HMOX1 were associated with prolonged respiratory failure, prolonged hospitalization, extensive airway necrosis, and CAS (P < 0.05). CONCLUSIONS These findings implicate donor bronchial hypoxia as a driving factor for post-transplantation airway complications. Strategies to improve airway oxygenation, such as bronchial artery re-anastomosis and hyperbaric oxygen therapy merit clinical investigation.
Collapse
Affiliation(s)
- Bryan D Kraft
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | | | - Eli C Colman
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Kamran Mahmood
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Matthew G Hartwig
- 3 Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Claude A Piantadosi
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,2 Department of Anesthesiology, and
| | - Scott L Shofer
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| |
Collapse
|
95
|
|
96
|
Angiogenesis in Ischemic Stroke and Angiogenic Effects of Chinese Herbal Medicine. J Clin Med 2016; 5:jcm5060056. [PMID: 27275837 PMCID: PMC4929411 DOI: 10.3390/jcm5060056] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 01/06/2023] Open
Abstract
Stroke is one of the major causes of death and adult disability worldwide. The underlying pathophysiology of stroke is highly complicated, consisting of impairments of multiple signalling pathways, and numerous pathological processes such as acidosis, glutamate excitotoxicity, calcium overload, cerebral inflammation and reactive oxygen species (ROS) generation. The current treatment for ischemic stroke is limited to thromolytics such as recombinant tissue plasminogen activator (tPA). tPA has a very narrow therapeutic window, making it suitable to only a minority of stroke patients. Hence, there is great urgency to develop new therapies that can protect brain tissue from ischemic damage. Recent studies have shown that new vessel formation after stroke not only replenishes blood flow to the ischemic area of the brain, but also promotes neurogenesis and improves neurological functions in both animal models and patients. Therefore, drugs that can promote angiogenesis after ischemic stroke can provide therapeutic benefits in stroke management. In this regard, Chinese herbal medicine (CHM) has a long history in treating stroke and the associated diseases. A number of studies have demonstrated the pro-angiogenic effects of various Chinese herbs and herbal formulations in both in vitro and in vivo settings. In this article, we present a comprehensive review of the current knowledge on angiogenesis in the context of ischemic stroke and discuss the potential use of CHM in stroke management through modulation of angiogenesis.
Collapse
|
97
|
Ohshima M, Yamaguchi Y, Ambe K, Horie M, Saito A, Nagase T, Nakashima K, Ohki H, Kawai T, Abiko Y, Micke P, Kappert K. Fibroblast VEGF-receptor 1 expression as molecular target in periodontitis. J Clin Periodontol 2016; 43:128-37. [PMID: 26932322 DOI: 10.1111/jcpe.12495] [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] [Accepted: 12/06/2015] [Indexed: 01/08/2023]
Abstract
AIM Degradation of extracellular matrices is an integral part in periodontitis. For antagonizing this pathophysiological mechanism, we aimed at identifying gene expression profiles in disease progression contributing periodontitis-associated fibroblasts (PAFs) versus normal gingival fibroblasts to determine their molecular repertoire, and exploit it for therapeutic intervention. MATERIALS AND METHODS Applying an exploratory analysis using a small number of microarrays in combination with a three dimensional (3D) in vitro culture model that incorporates some aspects of periodontitis, PAFs were initially characterized by gene-expression analyses, followed by targeted gene down-regulation and pharmacological intervention in vitro. Further, immunohistochemistry was applied for phosphorylation analyses in tissue specimens. RESULTS PAFs were characterized by 42 genes being commonly up-regulated >1.5-fold, and by five genes that were concordantly down-regulated (<0.7-fold). Expression of vascular endothelial growth factor (VEGF)-receptor 1 (Flt-1) was highly enhanced, and was thus further explored in in vitro culture models of periodontal fibroblasts without accounting for the microbiome. Phosphorylation of the VEGF-receptor 1 was enhanced in PAFs. Receptor inhibition by a specific VEGF-receptor inhibitor or intrinsic down-regulation by RNAi of the VEGF-receptor kinase in 3D gel cultures resulted in significant reduction in collagen degradation associated with increased tissue inhibitor of metalloproteinase expression, suggesting that Flt-1 may contribute to periodontitis. CONCLUSION Based on the finding that VEGF-receptor kinase inhibition impaired collagen degradation pathways, Flt-1 may represent a candidate for therapeutic approaches in periodontitis.
Collapse
Affiliation(s)
- Mitsuhiro Ohshima
- Department of Biochemistry, Ohu University School of Pharmaceutical Sciences, Koriyama, Fukushima, Japan
| | - Yoko Yamaguchi
- Department of Biochemistry, Nihon University School of Dentistry, Tokyo, Japan
| | - Kimiharu Ambe
- Department of Morphological Biology, Ohu University School of Dentistry, Koriyama, Fukushima, Japan
| | - Masafumi Horie
- Department of Respiratory Medicine, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Akira Saito
- Department of Respiratory Medicine, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Keisuke Nakashima
- Division of Periodontology, Department of Oral Function, Kyushu Dental University, Fukuoka, Japan
| | - Hidero Ohki
- First Department of Oral Surgery, Nihon University School of Dentistry, Tokyo, Japan
| | - Toshihisa Kawai
- Department of Immunology, The Forsyth Institute, Cambridge, MA, USA
| | - Yoshimitsu Abiko
- Department of Molecular Biology and Biochemistry, Nihon University School of Dentistry at Matsudo, Chiba, Japan
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Uppsala University, Hospital, Uppsala, Sweden
| | - Kai Kappert
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Center for Cardiovascular Research (CCR), Charité-University Medicine Berlin, Berlin, Germany
| |
Collapse
|
98
|
Olfert IM. Physiological Capillary Regression is not Dependent on Reducing VEGF Expression. Microcirculation 2016; 23:146-56. [PMID: 26660949 PMCID: PMC4744091 DOI: 10.1111/micc.12263] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/03/2015] [Indexed: 01/04/2023]
Abstract
Investigations into physiologically controlled capillary regression report the provocative finding that microvessel regression occurs in the face of persistent elevation of skeletal muscle VEGF expression. TSP-1, a negative angiogenic regulator, is increasingly being observed to temporally correlate with capillary regression, suggesting that increased TSP-1 (and not reduction in VEGF per se) is needed to initiate, and likely regulate, capillary regression. Based on evidence being gleaned from physiologically mediated regression of capillaries, it needs to be recognized that capillary regression (and perhaps capillary rarefaction with disease) is not simply the reversal of factors used to stimulate angiogenesis. Rather, the conceptual understanding that angiogenesis and capillary regression each have specific and unique requirements that are biologically constrained to opposite sides of the balance between positive and negative angioregulatory factors may shed light on why anti-VEGF therapies have not lived up to the promise in reversing angiogenesis and providing the cure that many had hoped toward fighting cancer. Emerging evidence from physiological controlled angiogenesis suggest that cases involving excessive or uncontrolled capillary expansion may be best treated by therapies designed to increase expression of negative angiogenic regulators, whereas those involving capillary rarefaction may benefit from inhibiting negative regulators (like TSP-1).
Collapse
Affiliation(s)
- I Mark Olfert
- Division of Exercise Physiology, Center for Cardiovascular and Respiratory Sciences, Mary Babb Randolph Cancer Center, West Virginia Clinical and Translational Science Institute, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| |
Collapse
|
99
|
Bikker A, Wielders J, van Loo R, Loubert M. Ascorbic acid deficiency impairs wound healing in surgical patients: Four case reports. INTERNATIONAL JOURNAL OF SURGERY OPEN 2016. [DOI: 10.1016/j.ijso.2016.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
100
|
Lee JS. Genetic Change from Colorectal Carcinoma Patients Using Comparative Genomic Hybridization. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2015. [DOI: 10.15324/kjcls.2015.47.4.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Jae Sik Lee
- Department of Clinical Laboratory Science, Hyejeon College, Hongseong 32244, Korea
| |
Collapse
|