1
|
Barron A, Tuulari J, Karlsson L, Karlsson H, O'Keeffe G, McCarthy C. Simulated ischaemia/reperfusion impairs trophoblast function through divergent oxidative stress- and MMP-9-dependent mechanisms. Biosci Rep 2024; 44:BSR20240763. [PMID: 39474810 PMCID: PMC11581840 DOI: 10.1042/bsr20240763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 11/22/2024] Open
Abstract
Early-onset pre-eclampsia is believed to arise from defective placentation in the first trimester, leading to placental ischaemia/reperfusion (I/R) and oxidative stress. However, our current understanding of the effects of I/R and oxidative stress on trophoblast function is ambiguous in part due to studies exposing trophoblasts to hypoxia instead of I/R, and which report conflicting results. Here, we present a model of simulated ischaemia/reperfusion (SI/R) to recapitulate the pathophysiological events of early-onset pre-eclampsia (PE), by exposing first trimester cytotrophoblast HTR-8/SVneo cells to a simulated ischaemia buffer followed by reperfusion. We examined different ischaemia and reperfusion times and observed that 1 h ischaemia and 24 h reperfusion induced an increase in reactive oxygen species (ROS) production (P<0.0001) and oxygen consumption rate (P<0.01). SI/R-exposed trophoblast cells exhibited deficits in migration, proliferation, and invasion (P<0.01). While the deficits in migration and proliferation were rescued by antioxidants, suggesting an ROS-dependent mechanism, the loss of invasion was not affected by antioxidants, which suggests a divergent ROS-independent pathway. In line with this, we observed a decrease in MMP-9, the key regulatory enzyme necessary for trophoblast invasion (P<0.01), which was similarly unaffected by antioxidants, and pharmacological inhibition of MMP-9 replicated the phenotype of deficient invasion (P<0.01). Collectively, these data demonstrate that I/R impairs trophoblast migration and proliferation via a ROS-dependent mechanism, and invasion via an ROS-independent loss of MMP-9, disambiguating the role of oxidative stress and providing insights into the response of trophoblasts to I/R in the context of early-onset PE.
Collapse
Affiliation(s)
- Aaron Barron
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Jetro J. Tuulari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychiatry and Turku Brain and Mind Centre, University of Turku and Turku University Hospital, Turku, Finland
- Turku Collegium for Science, Medicine and Technology, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku, Turku University Hospital, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku, Turku University Hospital, Turku, Finland
- Department of Clinical Medicine, Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychiatry and Turku Brain and Mind Centre, University of Turku and Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku, Turku University Hospital, Turku, Finland
| | - Gerard W. O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| |
Collapse
|
2
|
Wang Q, Yang J, Guo Z, Xi D. The predictive role of neutrophil gelatinase-associated lipocalin in coronary artery disease. Am J Med Sci 2024; 368:424-431. [PMID: 38906376 DOI: 10.1016/j.amjms.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
The prognosis holds significant implications for the long-term quality of life among patients suffering from coronary artery disease. However, a pressing challenge lies in the absence of reliable biomarkers that can establish a definitive correlation between these biomarkers and the prognosis of coronary artery heart disease. This review paper delves into the critical role of neutrophil gelatinase-associated lipocalin (NGAL) in predicting outcomes in coronary artery disease. It examines the influence of NGAL on various clinical manifestations, including stable angina, ST-segment elevation myocardial infarction, non-ST-segment elevation myocardial infarction, and isolated coronary artery dilation. Furthermore, this review provides recommendations aimed at enhancing the rigor and impact of future research, thereby serving as a valuable reference for subsequent studies in this domain.
Collapse
Affiliation(s)
- Qianqian Wang
- Department of Intensive Care Unit, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China; Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Junli Yang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Zhigang Guo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Dan Xi
- Department of Intensive Care Unit, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China.
| |
Collapse
|
3
|
Balasubramanian S, Perumal E. Integrated in silico analysis of transcriptomic alterations in nanoparticle toxicity across human and mouse models. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 949:174897. [PMID: 39053559 DOI: 10.1016/j.scitotenv.2024.174897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Nanoparticles, due to their exceptional physicochemical properties are used in our day-to-day environment. They are currently not regulated which might lead to increased levels in the biological systems causing adverse effects. However, the overall mechanism behind nanotoxicity remains elusive. Previously, we analysed the transcriptome datasets of copper oxide nanoparticles using in silico tools and identified IL-17, chemokine signaling pathway, and cytokine-cytokine receptor interaction as the key pathways altered. Based on the findings, we hypothesized a common pathway could be involved in transition metal oxide nanoparticles toxicity irrespective of the variables. Further, there could be unique transcriptome changes between metal oxide nanoparticles and other nanoparticles. To accomplish this, the overall transcriptome datasets of nanoparticles consisting of microarray and RNA-Seq were obtained. >90 studies for 17 different nanoparticles, performed in humans, rats, and mice were assessed. After initial screening, 24 mouse studies (with 196 datasets) and 34 human studies (with 200 datasets) were used for further analyses. The common genes that are dysregulated upon NPs exposure were identified for human and mouse datasets separately. Further, an overrepresentation functional enrichment analysis was performed. The common genes, their gene ontology, gene-gene, and protein-protein interactions were assessed. The overall results suggest that IL-17 and its related pathways might be commonly altered in nanoparticle exposure with lung as one of the major organs affected.
Collapse
Affiliation(s)
- Satheeswaran Balasubramanian
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India.
| |
Collapse
|
4
|
YUSTINASARI LR, KURATOMI M, KAGAWA S, GONDO A, ARAMAKI N, IMAI H, KUSAKABE KT. Specific expression and blood kinetics for relaxin 2, lipocalin 2, and tissue factor pathway inhibitor 2 at the canine placenta and pregnant bloods. J Vet Med Sci 2024; 86:77-86. [PMID: 38057091 PMCID: PMC10849861 DOI: 10.1292/jvms.23-0241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023] Open
Abstract
In general, humoral factors released from the placenta influence pregnancy progression, but the involvement of the canine placenta is often unidentified. We investigated specific genes in canine placentas and analyzed the blood dynamics of the translated proteins. Furthermore, RNAs are known to be released from placentas embedding in exosomes, a type of extracellular vesicles. Here, the presence of cell-free RNAs in pregnant serums was also confirmed. RNA specimens were purified from the normal healthy dog placentas and applied to RNA-Seq analysis. Expressions of frequent genes were confirmed by RT-PCR using placentas from other individuals and breeds. Relaxin (RLN) 2, lipocalin (LCN) 2, and tissue factor pathway inhibitor (TFPI) 2 were selected as high-expressed and placenta-specific genes. By western blot, the three factors were clearly detected in the pregnant serums. Quantitative analysis revealed that the amount of RLN2 increased significantly from non-pregnancy to day 41 of pregnancy. Regarding LCN2 and TFPI2, the protein serum levels elevated during pregnancy, but the statistical differences were not detected. Exosomes were found in all pregnant serums; however, the percentage was less than 6% in total extracellular vesicles. The cell-free RNA related to RLN2 was detected, but no elevation was confirmed during pregnancy. We found specific genes in the canine placenta and the transition of their translated protein into the blood. These factors may become useful tools for research on canine pregnancy and monitoring of reproductive management. Exosomes and cell-free RNA could not be found to be valid in canine reproduction.
Collapse
Affiliation(s)
- Lita Rakhma YUSTINASARI
- Laboratory of Basic Veterinary Science, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
- Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Maria KURATOMI
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Seizaburo KAGAWA
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Ai GONDO
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Nobuaki ARAMAKI
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Hiroyuki IMAI
- Laboratory of Basic Veterinary Science, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Ken Takeshi KUSAKABE
- Laboratory of Basic Veterinary Science, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
5
|
Kamiński M, Mierzyński R, Poniedziałek-Czajkowska E, Sadowska A, Sotowski M, Leszczyńska-Gorzelak B. Comparative Evaluation of Adipokine Metrics for the Diagnosis of Gestational Diabetes Mellitus. Int J Mol Sci 2023; 25:175. [PMID: 38203346 PMCID: PMC10778639 DOI: 10.3390/ijms25010175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common medical disorders in pregnancy. Adipokines, predominantly secreted by adipose tissue, are involved in numerous metabolic processes. The exact role of adipokines in the pathogenesis of GDM is still not well known, and numerous adipokines have been analysed throughout pregnancy and proposed as biomarkers of GDM. This study aimed to evaluate serum adiponectin, chemerin, lipocalin and apelin levels in GDM and non-GDM women, to assess them as clinically useful biomarkers of the occurrence of GDM and to demonstrate the correlation between the levels of the above adipokines in the blood serum and the increased risk of the development of GDM. The role of these adipokines in the pathogenesis of GDM was also analysed. The statistically significant differences between the levels of adiponectin (7234.6 vs. 9837.5 ng/mL, p < 0.0001), chemerin (264.0 vs. 206.7 ng/mL, p < 0.0001) and lipocalin (39.5 vs. 19.4 ng/mL, p < 0.0001) were observed between pregnant women with GDM and healthy ones. The diagnostic usefulness of the tested adipokines in detecting GDM was also assessed. The research results confirm the hypothesis on the significance of adiponectin, chemerin, lipocalin and apelin in the pathophysiological mechanisms of GDM. We speculate that these adipokines could potentially be established as novel biomarkers for the prediction and early diagnosis of GDM.
Collapse
Affiliation(s)
| | - Radzisław Mierzyński
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-954 Lublin, Poland; (M.K.); (A.S.); (M.S.); (B.L.-G.)
| | - Elżbieta Poniedziałek-Czajkowska
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-954 Lublin, Poland; (M.K.); (A.S.); (M.S.); (B.L.-G.)
| | | | | | | |
Collapse
|
6
|
Xia Q, Du Z, Chen M, Zhou X, Bai W, Zheng X, Lin L, Zhao Y, Ding J, Wu Z, Zou H, Wang S, Xu L, Li E, Wu B. A protein complex of LCN2, LOXL2 and MMP9 facilitates tumour metastasis in oesophageal cancer. Mol Oncol 2023; 17:2451-2471. [PMID: 37753805 PMCID: PMC10620126 DOI: 10.1002/1878-0261.13529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 08/21/2023] [Accepted: 09/25/2023] [Indexed: 09/28/2023] Open
Abstract
During malignant tumour development, the extracellular matrix (ECM) is usually abnormally regulated. Dysregulated expression of lysyl oxidase-like 2 (LOXL2), matrix metalloproteinase 9 (MMP9) and lipocalin 2 (LCN2) are associated with ECM remodelling. In this study, protein-protein interaction assays indicated that LCN2 and LOXL2 interactions and LCN2 and MMP9 interactions occurred both intracellularly and extracellularly, but interactions between LOXL2 and MMP9 only occurred intracellularly. The LCN2/LOXL2/MMP9 ternary complex promoted migration and invasion of oesophageal squamous cell carcinoma (ESCC) cells, as well as tumour growth and malignant progression in vivo, while the iron chelator deferoxamine mesylate (DFOM) inhibited ESCC tumour growth. Co-overexpression of LCN2, LOXL2 and MMP9 enhanced the ability of tumour cells to degrade fibronectin and Matrigel, increased the formation and extension of filopodia, and promoted the rearrangement of microfilaments through upregulation of profilin 1. In addition, the LCN2/LOXL2/MMP9 ternary complex promoted the expression of testican-1 (SPOCK1), and abnormally activated the FAK/AKT/GSK3β signalling pathway. In summary, the LCN2/LOXL2/MMP9 ternary complex promoted the migration and invasion of cancer cells and malignant tumour progression through multiple mechanisms and could be a potential therapeutic target.
Collapse
Affiliation(s)
- Qiaoxi Xia
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
- Central LaboratoryShantou Central HospitalChina
| | - Zepeng Du
- Central LaboratoryShantou Central HospitalChina
- Department of PathologyShantou Central HospitalChina
| | - Mantong Chen
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Xiao Zhou
- Central LaboratoryShantou Central HospitalChina
| | - Wenjing Bai
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Xiaoqi Zheng
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Ling Lin
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Yan Zhao
- Central LaboratoryShantou Central HospitalChina
| | - Jiyu Ding
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Zhisheng Wu
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Haiying Zou
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Shaohong Wang
- Department of PathologyShantou Central HospitalChina
| | - Liyan Xu
- Institute of Oncologic PathologyShantou University Medical CollegeChina
| | - Enmin Li
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Bingli Wu
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| |
Collapse
|
7
|
Suvakov S, Kattah AG, Gojkovic T, Enninga EAL, Pruett J, Jayachandran M, Sousa C, Santos J, Abou Hassan C, Gonzales-Suarez M, Garovic VD. Impact of Aging and Cellular Senescence in the Pathophysiology of Preeclampsia. Compr Physiol 2023; 13:5077-5114. [PMID: 37770190 DOI: 10.1002/cphy.c230003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The incidence of hypertensive disorders of pregnancy is increasing, which may be due to several factors, including an increased age at pregnancy and more comorbid health conditions during reproductive years. Preeclampsia, the most severe hypertensive disorder of pregnancy, has been associated with an increased risk of future disease, including cardiovascular and kidney diseases. Cellular senescence, the process of cell cycle arrest in response to many physiologic and maladaptive stimuli, may play an important role in the pathogenesis of preeclampsia and provide a mechanistic link to future disease. In this article, we will discuss the pathophysiology of preeclampsia, the many mechanisms of cellular senescence, evidence for the involvement of senescence in the development of preeclampsia, as well as evidence that cellular senescence may link preeclampsia to the risk of future disease. Lastly, we will explore how a better understanding of the role of cellular senescence in preeclampsia may lead to therapeutic trials. © 2023 American Physiological Society. Compr Physiol 13:5077-5114, 2023.
Collapse
Affiliation(s)
- Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea G Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Gojkovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth A L Enninga
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jacob Pruett
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ciria Sousa
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Janelle Santos
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
8
|
Li H, Konja D, Wang L, Wang Y. Sex Differences in Adiposity and Cardiovascular Diseases. Int J Mol Sci 2022; 23:ijms23169338. [PMID: 36012601 PMCID: PMC9409326 DOI: 10.3390/ijms23169338] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Body fat distribution is a well-established predictor of adverse medical outcomes, independent of overall adiposity. Studying body fat distribution sheds insights into the causes of obesity and provides valuable information about the development of various comorbidities. Compared to total adiposity, body fat distribution is more closely associated with risks of cardiovascular diseases. The present review specifically focuses on the sexual dimorphism in body fat distribution, the biological clues, as well as the genetic traits that are distinct from overall obesity. Understanding the sex determinations on body fat distribution and adiposity will aid in the improvement of the prevention and treatment of cardiovascular diseases (CVD).
Collapse
|
9
|
Neutrophil Gelatinase-Associated Lipocalin and Hypertensive Disorders of Pregnancy: A Cohort Study in Twin Pregnancies. J Clin Med 2022; 11:jcm11144163. [PMID: 35887927 PMCID: PMC9318711 DOI: 10.3390/jcm11144163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Abstract
Hypertensive disorders complicate more than 10% of twin pregnancies. Several studies showed increased neutrophil gelatinase-associated lipocalin (NGAL) values in women with singleton pregnancies and preeclampsia. This study aimed to assess NGAL values in twin pregnancies complicated by hypertensive disorders. We conducted a study of 242 consecutive twin pregnancies at the Medical University of Vienna. Serum NGAL was evaluated twice during pregnancy and once in the postpartum period. Furthermore, serum NGAL values were compared between women who developed hypertensive disorders and those who had normal blood pressure. In all twin pregnancies, mean NGAL values increased significantly from the first to the second visit (p = 0.004) and, further, after delivery (p < 0.001). NGAL was significantly higher in pregnancies that developed pregnancy hypertension or preeclampsia when compared to the control group at the first visit (109.2 ± 48.9 ng/mL vs. 91.9 ± 29.4 ng/mL, p = 0.04, respectively). The predictive power of first visit NGAL values for development of pregnancy hypertension or preeclampsia was evaluated. When using a cut-off value of 115 ng/mL, we obtained a sensitivity of 45% with a specificity of 77%. We conclude that women with twin pregnancies who develop hypertensive disorders of pregnancy showed increased NGAL values at 11−16 weeks.
Collapse
|
10
|
Dahal S, Dayal S, Androjna C, Peterson J, Ramamurthi A. Adult Mesenchymal Stem Cells and Derivatives in Improved Elastin Homeostasis in a Rat Model of Abdominal Aortic Aneurysms. Stem Cells Transl Med 2022; 11:850-860. [PMID: 35758561 PMCID: PMC9397656 DOI: 10.1093/stcltm/szac043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/04/2022] [Indexed: 12/02/2022] Open
Abstract
Abdominal aortic aneurysms (AAAs) are localized rupture-prone expansions of the aorta with limited reversibility that develop due to proteolysis of the elastic matrix. Natural regenerative repair of an elastic matrix is difficult due to the intrinsically poor elastogenicity of adult vascular smooth muscle cells (VSMCs). This justifies the need to provide external, pro-elastin regenerative- and anti-proteolytic stimuli to VSMCs in the AAA wall towards reinstating matrix structure in the aorta wall. Introducing alternative phenotypes of highly elastogenic and contractile cells into the AAA wall capable of providing such cues, proffers attractive prospects for AAA treatment. In this regard, we have previously demonstrated the superior elastogenicity of bone marrow mesenchymal stem cell (BM-MSC)-derived SMCs (cBM-SMCs) and their ability to provide pro-elastogenic and anti-proteolytic stimuli to aneurysmal SMCs in vitro. However, the major issues associated with cell therapy, such as their natural ability to home into the AAA tissue, their in vivo biodistribution and retention in the AAA wall, and possible paracrine effects on AAA tissue repair processes in the event of localization in remote tissues remain uncertain. Therefore, in this study we focused on assessing the fate, safety, and AAA reparative effects of BM-MSC-derived cBM-SMCs in vivo. Our results indicate that the cBM-SMCs (a) possess natural homing abilities similar to the undifferentiated BM-MSCs, (b) exhibit higher retention upon localization in the aneurysmal aorta than BM-MSCs, (c) downregulate the expression of several inflammatory and pro-apoptotic cytokines that are upregulated in the AAA wall contributing to accelerated elastic matrix breakdown and suppression of elastic fiber neo-assembly, repair, and crosslinking, and (d) improve elastic matrix content and structure in the AAA wall toward slowing the growth of AAAs. Our study provides initial evidence of the in vivo elastic matrix reparative benefits of cBM-SMCs and their utility in cell therapy to reverse the pathophysiology of AAAs.
Collapse
Affiliation(s)
- Shataakshi Dahal
- Lehigh University, Department of Bioengineering, Bethlehem, PA, USA
| | - Simran Dayal
- Lehigh University, Department of Bioengineering, Bethlehem, PA, USA
| | - Charlie Androjna
- Cleveland Clinic, Lerner Research Institute, Department of Biomedical Engineering, Cleveland, OH, USA
| | - John Peterson
- Cleveland Clinic, Lerner Research Institute, Department of Research Core Administration, Cleveland, OH, USA
| | - Anand Ramamurthi
- Lehigh University, Department of Bioengineering, Bethlehem, PA, USA
| |
Collapse
|
11
|
Lipocalin-2: Structure, function, distribution and role in metabolic disorders. Biomed Pharmacother 2021; 142:112002. [PMID: 34463264 DOI: 10.1016/j.biopha.2021.112002] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/26/2021] [Accepted: 08/01/2021] [Indexed: 12/27/2022] Open
Abstract
Lipocalin-2 (LCN-2) is a novel, 198 amino acid adipocytokine also referred to as neutrophil gelatinase-associated lipocalin (NGAL). LCN-2 is a circulatory protein responsible for the transportation of small and hydrophobic molecules (steroid, free fatty acids, prostaglandins and hormones) to target organs after binding to megalin/glycoprotein and GP330 SLC22A17 or 24p3R LCN-2 receptors. LCN-2 has been used as a biomarker for acute and chronic renal injury. It is present in a large variety of cells including neutrophil, hepatocytes, lung, bone marrow, adipose tissue, macrophages, thymus, non-neoplastic breast duct, prostate, and renal cells. Different functions have been associated with LCN-2. These functions include antibacterial, anti-inflammatory, and protection against cell and tissue stress. Moreover, LCN-2 can increase the pool of matrix metalloproteinase 9 in human neutrophil granulocytes. Other reported functions of LCN-2 include its ability to destroy the extracellular matrix, which could enable cancer progression and spread of metastasis. Recent reports show that the tissue level of LCN-2 is increased in metabolic disorders such as obesity and type 2 diabetes, suggesting an association between LCN-2 and insulin sensitivity and glucose homeostasis. The precise role of LCN-2 in the modulation of insulin sensitivity, glucose and lipid metabolism is still unclear. This review explores the structure of LCN-2, tissue distribution, and its interaction with important metabolic pathways.
Collapse
|
12
|
Dekens DW, Eisel ULM, Gouweleeuw L, Schoemaker RG, De Deyn PP, Naudé PJW. Lipocalin 2 as a link between ageing, risk factor conditions and age-related brain diseases. Ageing Res Rev 2021; 70:101414. [PMID: 34325073 DOI: 10.1016/j.arr.2021.101414] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022]
Abstract
Chronic (neuro)inflammation plays an important role in many age-related central nervous system (CNS) diseases, including Alzheimer's disease, Parkinson's disease and vascular dementia. Inflammation also characterizes many conditions that form a risk factor for these CNS disorders, such as physical inactivity, obesity and cardiovascular disease. Lipocalin 2 (Lcn2) is an inflammatory protein shown to be involved in different age-related CNS diseases, as well as risk factor conditions thereof. Lcn2 expression is increased in the periphery and the brain in different age-related CNS diseases and also their risk factor conditions. Experimental studies indicate that Lcn2 contributes to various neuropathophysiological processes of age-related CNS diseases, including exacerbated neuroinflammation, cell death and iron dysregulation, which may negatively impact cognitive function. We hypothesize that increased Lcn2 levels as a result of age-related risk factor conditions may sensitize the brain and increase the risk to develop age-related CNS diseases. In this review we first provide a comprehensive overview of the known functions of Lcn2, and its effects in the CNS. Subsequently, this review explores Lcn2 as a potential (neuro)inflammatory link between different risk factor conditions and the development of age-related CNS disorders. Altogether, evidence convincingly indicates Lcn2 as a key constituent in ageing and age-related brain diseases.
Collapse
Affiliation(s)
- Doortje W Dekens
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Leonie Gouweleeuw
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Regien G Schoemaker
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Peter P De Deyn
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Laboratory of Neurochemistry and Behaviour, Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Petrus J W Naudé
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands; Department of Psychiatry and Mental Health and Neuroscience Institute, Brain Behaviour Unit, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
13
|
Huang HL, Yang HL, Lai ZZ, Yang SL, Li MQ, Li DJ. Decidual IDO + macrophage promotes the proliferation and restricts the apoptosis of trophoblasts. J Reprod Immunol 2021; 148:103364. [PMID: 34482001 DOI: 10.1016/j.jri.2021.103364] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/14/2021] [Accepted: 08/18/2021] [Indexed: 12/29/2022]
Abstract
Indoleamine 2, 3-dioxygenase (IDO), a tryptophan-catabolizing enzyme, is essential in physiological immunoregulation. The present research was conducted to elucidate the expression and roles of IDO in decidual macrophages (dMφ) during early pregnancy. Here, we observed a remarkable decrease of IDO+ dMφ from patients with unexplained recurrent spontaneous abortion (URSA). IDO+ dMφ displayed M2 phenotype with higher CD206, CD209 and CD163, and lower CD86. Interestingly, treatment with 1-methyl-d-tryptophan (1-MT, an IDO pathway inhibitor) led to the M1 bias of dMφ. Further analysis of the cytokine array and the qPCR showed decreased levels of trophoblast proliferation or invasion-related molecules (e.g., CXCL12 and BMP2) in 1-MT-treated dMφ. The data of co-culture system showed that 1-MT-pretreated dMφ decreased the proliferation and the expression of Ki-67 and Bcl-2, and increased cell apoptosis of HTR-8/Snveo cells. Additionally, the expression of IDO in U937 cells was up-regulated by decidual stromal cells (DSC) and HTR-8/Snveo cells in vitro, as well as estradiol and medroxyprogesterone. These data suggest that endocrine environment, DSC and trophoblasts should contribute to the high level of IDO in dMφ, and IDO+ dMφ with M2 dominant phenotype promote the survival of trophoblasts during early pregnancy. The abnormal lower level of IDO should trigger the dysfunction of dMφ, further suppress the survival of trophoblasts and increase the risk of miscarriage.
Collapse
Affiliation(s)
- Hong-Lan Huang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China
| | - Hui-Li Yang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China
| | - Zhen-Zhen Lai
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China
| | - Shao-Liang Yang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China.
| | - Da-Jin Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
14
|
Colson A, Sonveaux P, Debiève F, Sferruzzi-Perri AN. Adaptations of the human placenta to hypoxia: opportunities for interventions in fetal growth restriction. Hum Reprod Update 2020; 27:531-569. [PMID: 33377492 DOI: 10.1093/humupd/dmaa053] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The placenta is the functional interface between the mother and the fetus during pregnancy, and a critical determinant of fetal growth and life-long health. In the first trimester, it develops under a low-oxygen environment, which is essential for the conceptus who has little defense against reactive oxygen species produced during oxidative metabolism. However, failure of invasive trophoblasts to sufficiently remodel uterine arteries toward dilated vessels by the end of the first trimester can lead to reduced/intermittent blood flow, persistent hypoxia and oxidative stress in the placenta with consequences for fetal growth. Fetal growth restriction (FGR) is observed in ∼10% of pregnancies and is frequently seen in association with other pregnancy complications, such as preeclampsia (PE). FGR is one of the main challenges for obstetricians and pediatricians, as smaller fetuses have greater perinatal risks of morbidity and mortality and postnatal risks of neurodevelopmental and cardio-metabolic disorders. OBJECTIVE AND RATIONALE The aim of this review was to examine the importance of placental responses to changing oxygen environments during abnormal pregnancy in terms of cellular, molecular and functional changes in order to highlight new therapeutic pathways, and to pinpoint approaches aimed at enhancing oxygen supply and/or mitigating oxidative stress in the placenta as a mean of optimizing fetal growth. SEARCH METHODS An extensive online search of peer-reviewed articles using PubMed was performed with combinations of search terms including pregnancy, placenta, trophoblast, oxygen, hypoxia, high altitude, FGR and PE (last updated in May 2020). OUTCOMES Trophoblast differentiation and placental establishment are governed by oxygen availability/hypoxia in early pregnancy. The placental response to late gestational hypoxia includes changes in syncytialization, mitochondrial functions, endoplasmic reticulum stress, hormone production, nutrient handling and angiogenic factor secretion. The nature of these changes depends on the extent of hypoxia, with some responses appearing adaptive and others appearing detrimental to the placental support of fetal growth. Emerging approaches that aim to increase placental oxygen supply and/or reduce the impacts of excessive oxidative stress are promising for their potential to prevent/treat FGR. WIDER IMPLICATIONS There are many risks and challenges of intervening during pregnancy that must be considered. The establishment of human trophoblast stem cell lines and organoids will allow further mechanistic studies of the effects of hypoxia and may lead to advanced screening of drugs for use in pregnancies complicated by placental insufficiency/hypoxia. Since no treatments are currently available, a better understanding of placental adaptations to hypoxia would help to develop therapies or repurpose drugs to optimize placental function and fetal growth, with life-long benefits to human health.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
15
|
Camacho CA, Santos GDO, Caballeros JE, Cazales N, Ramirez CJ, Vidigal PMP, Ramos HJDO, Barros E, Mattos RC. Uterine infusion of conceptus fragments changes the protein profile from cyclic mares. Anim Reprod 2020; 17:e20200552. [PMID: 33791032 PMCID: PMC7995263 DOI: 10.1590/1984-3143-ar2020-0552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 11/22/2022] Open
Abstract
This experiment aimed to compare at day seven after ovulation, the protein profile of uterine fluid in cyclic mares with mares infused two days before with Day 13 conceptus fragments. Experimental animals were ten healthy cyclic mares, examined daily to detect ovulation (Day 0) as soon as estrus was confirmed. On day seven, after ovulation, uterine fluid was collected, constituting the Cyclic group (n = 10). The same mares were examined in the second cycle until ovulation was detected. On day five, after ovulation, fragments from a previously collected concepti were infused into each mare's uterus. Two days after infusion, uterine fluid was collected, constituting the Fragment group (n = 10). Two-dimensional electrophoresis technique processed uterine fluid samples. A total of 373 spots were detected. MALDI-TOF/TOF and NanoUHPLC-QTOF mass spectrometry identified twenty spots with differences in abundance between the Cyclic and Fragment group. Thirteen proteins were identified, with different abundance between groups. Identified proteins may be related to embryo-maternal communication, which involves adhesion, nutrition, endothelial cell proliferation, transport, and immunological tolerance. In conclusion, conceptus fragments signalized changes in the protein profile of uterine fluid seven days after ovulation in comparison to the observed at Day 7 in the same cyclic mares.
Collapse
Affiliation(s)
- Cesar Augusto Camacho
- Laboratório de Reprodução Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Gabriel de Oliveira Santos
- Laboratório de Reprodução Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Jorge Emilio Caballeros
- Laboratório de Reprodução Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Nicolas Cazales
- Laboratório de Reprodução Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
- Facultad de Veterinária, Universidad de la República - UDELAR, Montevideo, Uruguay
| | - Camilo José Ramirez
- Núcleo de Análise de Biomoléculas, Universidade Federal de Viçosa, Viçosa, MG, Brasil
| | | | | | - Edvaldo Barros
- Núcleo de Análise de Biomoléculas, Universidade Federal de Viçosa, Viçosa, MG, Brasil
| | - Rodrigo Costa Mattos
- Laboratório de Reprodução Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| |
Collapse
|
16
|
Zhang I, Lépine P, Han C, Lacalle-Aurioles M, Chen CXQ, Haag R, Durcan TM, Maysinger D. Nanotherapeutic Modulation of Human Neural Cells and Glioblastoma in Organoids and Monocultures. Cells 2020; 9:cells9112434. [PMID: 33171886 PMCID: PMC7695149 DOI: 10.3390/cells9112434] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammatory processes in the brain are orchestrated by microglia and astrocytes in response to activators such as pathogen-associated molecular patterns, danger-associated molecular patterns and some nanostructures. Microglia are the primary immune responders in the brain and initiate responses amplified by astrocytes through intercellular signaling. Intercellular communication between neural cells can be studied in cerebral organoids, co-cultures or in vivo. We used human cerebral organoids and glioblastoma co-cultures to study glia modulation by dendritic polyglycerol sulfate (dPGS). dPGS is an extensively studied nanostructure with inherent anti-inflammatory properties. Under inflammatory conditions, lipocalin-2 levels in astrocytes are markedly increased and indirectly enhanced by soluble factors released from hyperactive microglia. dPGS is an effective anti-inflammatory modulator of these markers. Our results show that dPGS can enter neural cells in cerebral organoids and glial cells in monocultures in a time-dependent manner. dPGS markedly reduces lipocalin-2 abundance in the neural cells. Glioblastoma tumoroids of astrocytic origin respond to activated microglia with enhanced invasiveness, whereas conditioned media from dPGS-treated microglia reduce tumoroid invasiveness. Considering that many nanostructures have only been tested in cancer cells and rodent models, experiments in human 3D cerebral organoids and co-cultures are complementary in vitro models to evaluate nanotherapeutics in the pre-clinical setting. Thoroughly characterized organoids and standardized procedures for their preparation are prerequisites to gain information of translational value in nanomedicine. This study provides data for a well-characterized dendrimer (dPGS) that modulates the activation state of human microglia implicated in brain tumor invasiveness.
Collapse
Affiliation(s)
- Issan Zhang
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada;
| | - Paula Lépine
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Chanshuai Han
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - María Lacalle-Aurioles
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Carol X.-Q. Chen
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany;
| | - Thomas M. Durcan
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada;
- Correspondence: ; Tel.: +1-514-398-1264
| |
Collapse
|
17
|
Yamada Y, Miyamoto T, Higuchi S, Ono M, Kobara H, Asaka R, Ando H, Suzuki A, Shiozawa T. cDNA expression library screening revealed novel functional genes involved in clear cell carcinogenesis of the ovary in vitro. J OBSTET GYNAECOL 2020; 41:100-105. [PMID: 32157937 DOI: 10.1080/01443615.2020.1716310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In order to identify genes involved in the pathogenesis of clear cell carcinoma of the ovary (CCC), functional screening using a cDNA expression library was performed. We extracted mRNA from a CCC cell line (RMG-1), established a cDNA library using a retroviral vector, transfected that library into mouse NIH3T3 cells and sequenced the resultant foci. The tissue-type specific expression of isolated genes and their transforming activities were evaluated. Seven genes were isolated. Of these genes, the mRNA expression of SEC61B and DVL1 is significantly stronger in CCC than in other histological types (p < .05). Immunohistochemical staining reveals the stronger expression of SEC61B and C1ORF38 than normal ovarian tissues (p < .05). Focus formation is confirmed by the transfection of SEC61B, C1ORF38, and DVL1 into NIH3T3 cells. The present study identified novel genes including SEC61B, C1ORF38, and DVL1, involved in the pathogenesis of CCC. These genes may be additional therapeutic targets for CCC.Impact statementWhat is already known on this subject? Several important genetic abnormalities, including ARID1A and PIK3CA mutations, have been reported in ovarian clear cell carcinoma (CCC).What the results of this study add? SEC61B, C1ORF38, and DVL1 were newly detected as candidate genes involved in ovarian clear cell carcinogenesis.What the implications are of these findings for clinical practice and/or further research? Functional screening using a cDNA expression library may be a useful technique to identify functional genes for pathogenesis. The information obtained using this technique may provide new therapeutic targets of CCC.
Collapse
Affiliation(s)
- Yasushi Yamada
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Tsutomu Miyamoto
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Shotaro Higuchi
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Motoki Ono
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Hisanori Kobara
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Ryoichi Asaka
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Hirofumi Ando
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Akihisa Suzuki
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Tanri Shiozawa
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| |
Collapse
|
18
|
Yin X, Huo Y, Liu L, Pan Y, Liu S, Wang R. Serum Levels and Placental Expression of NGAL in Gestational Diabetes Mellitus. Int J Endocrinol 2020; 2020:8760563. [PMID: 32377189 PMCID: PMC7199580 DOI: 10.1155/2020/8760563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/17/2019] [Accepted: 12/09/2019] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES The aim was to investigate neutrophil gelatinase-associated lipocalin (NGAL) levels in the serum and term placentas and its potential role in gestational diabetes mellitus (GDM). METHODS A total of 49 GDM subjects and 39 age-matched women with normal pregnancies were recruited. We examined serum concentrations of NGAL and tumor necrosis factor-α (TNF-α) in maternal blood and cord blood and their expression levels in the term placentas and umbilical cord. RESULTS Serum NGAL levels were significantly higher in GDM patients than in normal pregnant controls both in the maternal blood (4.80 ± 1.99 vs. 3.66 ± 1.13, P=0.001) and the cord blood (4.70 ± 2.08 vs. 3.85 ± 1.44, P=0.027). Moreover, serum NGAL levels exhibited a positive correlation with various parameters of insulin resistance. Maternal serum NGAL levels positively correlated with the NGAL levels found in the cord blood of the control (r = 0.399, P=0.012) and the GDM subjects (r = 0.349, P=0.014). Finally, the expression of NGAL protein levels in the placenta (1.22 ± 0.39 vs. 0.65 ± 0.23, P < 0.001) and umbilical cord (0.65 ± 0.23 vs. 0.25 ± 0.10, P < 0.001) were higher in GDM women than those noted in the control subjects. In the GDM group, maternal serum NGAL levels exhibited a positive correlation with placental NGAL mRNA and protein levels (r = 0.848, P=0.008; r = 0.636, P=0.011, respectively). CONCLUSIONS NGAL may be an important adipokine involved in GDM and fetal development. The oversecretion of NGAL from the placenta may contribute to the elevated levels of serum NGAL in gestational diabetes mellitus.
Collapse
Affiliation(s)
- Xiaoqian Yin
- Department of Obstetrics & Gynecology, Hebei General Hospital, Shijiazhuang 050051, China
| | - Yan Huo
- Department of Obstetrics & Gynecology, Hebei General Hospital, Shijiazhuang 050051, China
| | - Li Liu
- Department of Obstetrics & Gynecology, Hebei General Hospital, Shijiazhuang 050051, China
| | - Yixing Pan
- Department of Obstetrics & Gynecology, Hebei General Hospital, Shijiazhuang 050051, China
| | - Suxin Liu
- Department of Obstetrics & Gynecology, Hebei General Hospital, Shijiazhuang 050051, China
| | - Runfang Wang
- Department of Obstetrics & Gynecology, Hebei General Hospital, Shijiazhuang 050051, China
| |
Collapse
|
19
|
Schumacher A, Zenclussen AC. Human Chorionic Gonadotropin-Mediated Immune Responses That Facilitate Embryo Implantation and Placentation. Front Immunol 2019; 10:2896. [PMID: 31921157 PMCID: PMC6914810 DOI: 10.3389/fimmu.2019.02896] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022] Open
Abstract
Human chorionic gonadotropin (hCG) serves as one of the first signals provided by the embryo to the mother. Exactly at the time when the first step of the implantation process is initiated and the blastocyst adheres to the maternal endometrium, the embryonic tissue starts to actively secrete hCG. Shortly thereafter, the hormone can be detected in the maternal circulation where its concentration steadily increases throughout early pregnancy as it is continuously released by the forming placenta. Accumulating evidence underlines the critical function of hCG for embryo implantation and placentation. hCG not only regulates biological aspects of these early pregnancy events but also supports maternal immune cells in their function as helpers in the establishment of an adequate embryo-endometrial relationship. In view of its early presence in the maternal circulation, hCG has the potential to influence both local uterine immune cell populations as well as peripheral ones. The current review aims to summarize recent literature on the participation of innate and adaptive immune cells in embryo implantation and placentation with a specific focus on their regulation by hCG.
Collapse
Affiliation(s)
- Anne Schumacher
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ana C Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
20
|
Lei D, Deng N, Wang S, Huang J, Fan C. Upregulated ARRDC3 limits trophoblast cell invasion and tube formation and is associated with preeclampsia. Placenta 2019; 89:10-19. [PMID: 31665660 DOI: 10.1016/j.placenta.2019.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 10/12/2019] [Accepted: 10/15/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Bioinformatics analysis indicated that the arrestin ARRDC3 was upregulated in placental tissue from patients with preeclampsia (PE). The study aimed to confirm the finding by examining placenta samples from women with and without early-onset PE and to investigate ARRDC3 roles in trophoblast function. METHODS ARRDC3 expression level and localization in placental tissue were determined by Western blot, real-time quantitative PCR and immunohistochemistry. An in vitro hypoxia and an in vitro ischemia (hypoxia/reoxygenation) cell models were used to determine the hypoxic and ischemic effects on ARRDC3 expression in extravillous trophoblast-derived HTR/8SVneo cells and trophoblast cell activity. The role of ARRDC3 in HTR8/SVneo cell proliferation, invasion and tube formation in vitro was investigated by testing the effects of ARRDC3 gene overexpression or siRNA-based gene silencing. RESULTS ARRDC3 expression was significantly elevated in placental tissue from women with early-onset PE compared to preterm birth pregnancies. ARRDC3 protein was localized in human placental trophoblasts. Hypoxia and ischemia both enhanced ARRDC3 protein expression in HTR8/SVneo cells. Hypoxia altered trophoblast cell activities. Overexpression of ARRDC3 in HTR8/SVneo cells suppressed cell invasion and tube formation. ARRDC3 gene silencing, by contrast, promoted invasion and tube formation under hypoxic conditions. CONCLUSION ARRDC3 was highly expressed in placental tissues of PE patients and directly affected biological activities of trophoblasts under hypoxic conditions. In regulation of ARRDC3- protein expression, ischemia (hypoxia/reoxygenation) are also important. These findings suggest that ARRDC3 may play a clinically significant role in the pathogenesis of PE.
Collapse
Affiliation(s)
- Di Lei
- Department of Obstetrics and Gynecology, Renmin Hospital, Wuhan University, Hubei, PR China
| | - Na Deng
- Department of Obstetrics and Gynecology, Renmin Hospital, Wuhan University, Hubei, PR China
| | - Suqing Wang
- Department of Preventive Medicine, School of Health Science, Wuhan University, Wuhan, Hubei, PR China
| | - Jinfa Huang
- Department of Obstetrics and Gynecology, Renmin Hospital, Wuhan University, Hubei, PR China
| | - Cuifang Fan
- Department of Obstetrics and Gynecology, Renmin Hospital, Wuhan University, Hubei, PR China.
| |
Collapse
|
21
|
Eirini Papathanasiou A, Malamitsi-Puchner A, Gavrili S, Zachaki S, Georgantzi S, Marmarinos A, Christou C, Voulgaris K, Gourgiotis D, Briana DD. Perinatal lipocalin-2 profile at the extremes of fetal growth. J Matern Fetal Neonatal Med 2019; 34:2166-2172. [PMID: 31450995 DOI: 10.1080/14767058.2019.1659774] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Lipocalin-2 (LCN-2) has been identified as an osteoblast-secreted hormone regulating immunity, inflammation and metabolic homeostasis and has emerged as a diagnostic and prognostic biomarker for acute kidney injury in neonates. We investigated the impact of fetal growth on antepartum maternal serum, cord serum and breast milk LCN-2 concentrations and the associations of the latter with perinatal parameters. METHODS Maternal serum, cord serum and breast milk LCN-2 concentrations were measured by ELISA in samples from 80 mothers who delivered 40 appropriate (AGA), 20 large for gestational age (LGA) and 20 intrauterine growth restricted (IUGR) neonates, classified by customized weight centiles. LCN-2 concentrations were associated with birth weight, customized centile, gender, maternal age and delivery mode. RESULTS Antepartum maternal serum LCN-2 concentrations were significantly higher in women delivering AGA infants compared to the other two groups. Cord blood LCN-2 concentrations were significantly higher compared to maternal ones; furthermore, they were significantly elevated in the IUGR group compared to the LGA one (p = .019). Lowest concentrations were detected in breast milk, which did not differ between the three growth groups. A negative correlation was documented between cord blood LCN-2 concentrations and customized centiles (r: -0.304, p = .007). CONCLUSIONS The higher cord serum LCN-2 concentrations, compared to maternal ones, may point to its fetal origin and potential role in intrauterine growth. The negative correlation of cord LCN-2 concentrations with customized centiles, possibly implies reduced nephron endowment/subclinical kidney damage in IUGR neonates. The extremely low LCN-2 breast milk concentrations could imply that the secretion of LCN-2 from maternal circulation to breast milk is not influenced by factors leading to intrauterine growth pathology.
Collapse
Affiliation(s)
| | | | - Stavroula Gavrili
- Neonatal Intensive Care Unit, "Alexandra" University and State Maternity Hospital, Athens, Greece
| | - Sophia Zachaki
- Laboratory of Health Physics, Radiobiology and Cytogenetics, National Center for Scientific Research (NCSR) "Demokritos", Athens, Greece
| | - Sophia Georgantzi
- Neonatal Intensive Care Unit, "Alexandra" University and State Maternity Hospital, Athens, Greece
| | - Antonios Marmarinos
- Laboratory of Clinical Biochemistry - Molecular Diagnostics, 2nd Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Dimitrios Gourgiotis
- Laboratory of Clinical Biochemistry - Molecular Diagnostics, 2nd Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina D Briana
- Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
22
|
Higuchi S, Miyamoto T, Kobara H, Yamada S, Asaka R, Kikuchi N, Kashima H, Ohira S, Shiozawa T. Trophoblast type-specific expression of senescence markers in the human placenta. Placenta 2019; 85:56-62. [DOI: 10.1016/j.placenta.2019.06.377] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 05/16/2019] [Accepted: 06/21/2019] [Indexed: 01/05/2023]
|
23
|
Olmos-Ortiz A, Flores-Espinosa P, Mancilla-Herrera I, Vega-Sánchez R, Díaz L, Zaga-Clavellina V. Innate Immune Cells and Toll-like Receptor-Dependent Responses at the Maternal-Fetal Interface. Int J Mol Sci 2019; 20:ijms20153654. [PMID: 31357391 PMCID: PMC6695670 DOI: 10.3390/ijms20153654] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022] Open
Abstract
During pregnancy, the placenta, the mother and the fetus exploit several mechanisms in order to avoid fetal rejection and to maintain an immunotolerant environment throughout nine months. During this time, immune cells from the fetal and maternal compartments interact to provide an adequate defense in case of an infection and to promote a tolerogenic milieu for the fetus to develop peacefully. Trophoblasts and decidual cells, together with resident natural killer cells, dendritic cells, Hofbauer cells and other macrophages, among other cell types, contribute to the modulation of the uterine environment to sustain a successful pregnancy. In this review, the authors outlined some of the various roles that the innate immune system plays at the maternal-fetal interface. First, the cell populations that are recruited into gestational tissues and their immune mechanisms were examined. In the second part, the Toll-like receptor (TLR)-dependent immune responses at the maternal-fetal interface was summarized, in terms of their specific cytokine/chemokine/antimicrobial peptide expression profiles throughout pregnancy.
Collapse
Affiliation(s)
- Andrea Olmos-Ortiz
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México 11000, Mexico
| | - Pilar Flores-Espinosa
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México 11000, Mexico
| | - Ismael Mancilla-Herrera
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México 11000, Mexico
| | - Rodrigo Vega-Sánchez
- Departamento de Nutrición y Bioprogramación, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México 11000, Mexico
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Verónica Zaga-Clavellina
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México 11000, Mexico.
| |
Collapse
|
24
|
Chaudhary P, Babu GS, Sobti RC, Gupta SK. HGF regulate HTR-8/SVneo trophoblastic cells migration/invasion under hypoxic conditions through increased HIF-1α expression via MAPK and PI3K pathways. J Cell Commun Signal 2019; 13:503-521. [PMID: 30684191 DOI: 10.1007/s12079-019-00505-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/17/2019] [Indexed: 12/22/2022] Open
Abstract
Hepatocyte growth factor (HGF) is reported to be down-regulated in pregnancy complications like intrauterine growth retardation and preeclampsia, which are associated with abnormal trophoblast migration/invasion. In this study, role of HGF and associated signaling pathways has been investigated in HTR-8/SVneo trophoblastic cells migration/invasion under normoxia (20% O2) and hypoxia (2% O2). HTR-8/SVneo cells exposed to hypoxia showed increase in migration and invasion as compared to cells incubated under normoxic conditions. The migration/invasion under both normoxic and hypoxic conditions was further enhanced after treatment with HGF. Subsequent to treatment with HGF, a significant increase in expression of MMP2 & MMP3 under normoxia and MMP1 & MMP9 under hypoxia was observed. Treatment of HTR-8/SVneo cells with HGF under hypoxia also led to decrease in TIMP1. Treatment of the cells with HGF led to activation of mitogen activated protein kinases (MAPK) and phosphatidylinositol 3-kinase (PI3K) signaling pathways. Inhibition of MAPK by U0126 and PI3K by LY294002 led to concomitant decrease in the HGF-mediated migration/invasion of HTR-8/SVneo cells. HGF treatment under hypoxia also led to a significant increase in hypoxia inducible factor (HIF-1α) expression. Additionally, inhibition of HIF-1α by siRNA led to decrease in HGF-mediated migration of HTR-8/SVneo cells under hypoxic conditions. Inhibition of HGF activated MAPK and PI3K signaling led to reduction in HIF-1α expression under hypoxia. In conclusion, HGF facilitates HTR-8/SVneo cell migration/invasion by activation of MAPK/PI3K signaling pathways and increased expression of MMPs. HIF-1α has a role in HGF-mediated increase in migration under hypoxic conditions.
Collapse
Affiliation(s)
- Piyush Chaudhary
- Reproductive Cell Biology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India.,Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Lucknow, 226 025, India
| | - Gosipatala Sunil Babu
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Lucknow, 226 025, India
| | - Ranbir Chander Sobti
- Department of Biotechnology, Panjab University, Sector-14, Chandigarh, 160 014, India
| | - Satish Kumar Gupta
- Reproductive Cell Biology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India.
| |
Collapse
|
25
|
Choudhury RH, Dunk CE, Lye SJ, Harris LK, Aplin JD, Jones RL. Decidual leucocytes infiltrating human spiral arterioles are rich source of matrix metalloproteinases and degrade extracellular matrix in vitro and in situ. Am J Reprod Immunol 2019; 81:e13054. [PMID: 30267451 DOI: 10.1111/aji.13054] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 01/02/2023] Open
Abstract
PROBLEM During pregnancy, the decidual spiral arterioles (SpAs) that supply maternal blood to the placenta undergo a series of changes to optimise the transfer of nutrients and oxygen to the developing foetus. Recent studies have shown that initiation of SpA transformation coincides with decidual leucocyte infiltration. Leucocytes are known to be a source of matrix metalloproteinases (MMPs); however, the complete profile of MMPs expressed by decidual NK cells (dNK) and macrophages has not been characterised. We hypothesised that leucocyte-derived MMPs contribute to SpA remodelling. METHODS Decidual NK cells and macrophages were isolated from first trimester decidua and their MMP repertoire profiled by qRT-PCR (n = 10; 5-11 weeks). Dual immunofluorescence was used to localise MMP expression in situ (n = 3; 5-12 weeks). Gelatin zymography was carried out to assess whether leucocyte-derived MMPs can degrade ECM. In situ zymography and immunofluorescence identified MMP activity in tissue-resident dNK and macrophages. RESULTS Decidual NK cells cells and macrophages expressed MMP2, -7, -9, -11, -16, -19 and tissue inhibitors of metalloproteinase-1, -2, and -3. Both cell types degraded gelatin using MMP2 and MMP9 and broke down collagen in an in vitro model of the SpA. Extravillous trophoblasts (EVTs) expressed a similar repertoire of MMPs. CONCLUSION We suggest that matrix remodelling in SpA is initiated by infiltrating leucocytes, while EVTs become involved at later stages.
Collapse
Affiliation(s)
- Ruhul H Choudhury
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester, UK
- Academic Health Science Centre, St Mary's Hospital, Manchester, UK
| | - Caroline E Dunk
- Research Centre for Women's and Infants' Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Stephen J Lye
- Research Centre for Women's and Infants' Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Lynda K Harris
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester, UK
- Academic Health Science Centre, St Mary's Hospital, Manchester, UK
- Manchester Pharmacy School, University of Manchester, Manchester, UK
| | - John D Aplin
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester, UK
- Academic Health Science Centre, St Mary's Hospital, Manchester, UK
| | - Rebecca L Jones
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester, UK
- Academic Health Science Centre, St Mary's Hospital, Manchester, UK
| |
Collapse
|
26
|
Miyamoto T, Kashima H, Yamada Y, Kobara H, Asaka R, Ando H, Higuchi S, Ida K, Mvunta DH, Shiozawa T. Lipocalin 2 Enhances Migration and Resistance against Cisplatin in Endometrial Carcinoma Cells. PLoS One 2016; 11:e0155220. [PMID: 27168162 PMCID: PMC4864227 DOI: 10.1371/journal.pone.0155220] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 04/26/2016] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Lipocalin 2 (LCN2) is a secretory protein that is involved in various physiological processes including iron transport. We previously identified LCN2 as an up-regulated gene in endometrial carcinoma, and found that the overexpression of LCN2 and its receptor, SLC22A17, was associated with a poor prognosis. However, the functions and mechanism of action of LCN2 currently remain unclear. METHODS The LCN2-overexpressing endometrial carcinoma cell lines, HHUA and RL95-2, and LCN2-low-expressing one, HEC1B, were used. The effects of LCN2 on cell migration, cell viability, and apoptosis under various stresses, including ultraviolet (UV) irradiation and cisplatin treatment, were examined using the scratch wound healing assay, WST-1 assay, and Apostrand assay, respectively. RESULTS LCN2-silencing using shRNA method significantly reduced the migration ability of cells (p<0.05). Cytotoxic stresses significantly decreased the viability of LCN2-silenced cells more than that of control cells. In contrast, LCN2 overexpression was significantly increased cisplatin resistance. These effects were canceled by the addition of the iron chelator, deferoxamine. After UV irradiation, the expression of phosphorylated Akt (pAkt) was decreased in LCN2-silenced cells, and the PI3K inhibitor canceled the difference induced in UV sensitivity by LCN2. The cisplatin-induced expression of pAkt was not affected by LCN2; however, the expression of p53 and p21 was increased by LCN2-silencing. CONCLUSIONS These results indicated that LCN2 was involved in the migration and survival of endometrial carcinoma cells under various stresses in an iron-dependent manner. The survival function of LCN2 may be exerted through the PI3K pathway and suppression of the p53-p21 pathway. These functions of LCN2 may increase the malignant potential of endometrial carcinoma cells.
Collapse
Affiliation(s)
- Tsutomu Miyamoto
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
- * E-mail:
| | - Hiroyasu Kashima
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Yasushi Yamada
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Hisanori Kobara
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Ryoichi Asaka
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Hirofumi Ando
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Shotaro Higuchi
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Koichi Ida
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - David Hamisi Mvunta
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Tanri Shiozawa
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| |
Collapse
|
27
|
Xia C, Li XQ, Zhou ZH, Chen HS. Identification of cytokines for early prediction of malignant middle cerebral artery infarction. Int J Neurosci 2016; 127:86-91. [DOI: 10.3109/00207454.2016.1146265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
28
|
Yamada Y, Miyamoto T, Kashima H, Kobara H, Asaka R, Ando H, Higuchi S, Ida K, Shiozawa T. Lipocalin 2 attenuates iron-related oxidative stress and prolongs the survival of ovarian clear cell carcinoma cells by up-regulating the CD44 variant. Free Radic Res 2016; 50:414-25. [PMID: 26729415 DOI: 10.3109/10715762.2015.1134795] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Ovarian clear cell carcinoma (CCC) arises from ovarian endometriosis. Intra-cystic fluid contains abundant amounts of free iron, which causes persistent oxidative stress, a factor that has been suggested to induce malignant transformation. However, the mechanisms linking oxidative stress and carcinogenesis in CCC currently remain unclear. Lipocalin 2 (LCN2), a multifunctional secretory protein, functions as an iron transporter as well as an antioxidant. Therefore, we herein examined the roles of LCN2 in the regulation of intracellular iron concentrations, oxidative stress, DNA damage, and antioxidative functions using LCN2-overexpressing (ES2), and LCN2-silenced (RMG-1) CCC cell lines. The results of calcein staining indicated that the up-regulated expression of LCN2 correlated with increases in intracellular iron concentrations. However, a DCFH-DA assay and 8OHdG staining revealed that LCN2 reduced intracellular levels of reactive oxygen species and DNA damage. Furthermore, the expression of LCN2 suppressed hydrogen peroxide-induced apoptosis and prolonged cell survival, suggesting an antioxidative role for LCN2. The expression of mRNAs and proteins for various oxidative stress-catalyzing enzymes, such as heme oxygenase (HO), superoxide dismutase (SOD), and glutathione peroxidase, was not affected by LCN2, whereas the intracellular concentration of the potent antioxidant, glutathione (GSH), was increased by LCN2. Furthermore, the expression of xCT, a cystine transporter protein, and CD44 variant 8-10 (CD44v), a stem cell marker, was up-regulated by LCN2. Although LCN2 increased intracellular iron concentrations, LCN2-induced GSH may catalyze and override oxidative stress via CD44v and xCT, and subsequently enhance the survival of CCC cells in oxidative stress-rich endometriosis.
Collapse
Affiliation(s)
- Yasushi Yamada
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Tsutomu Miyamoto
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Hiroyasu Kashima
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Hisanori Kobara
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Ryoichi Asaka
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Hirofumi Ando
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Shotaro Higuchi
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Koichi Ida
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Tanri Shiozawa
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| |
Collapse
|
29
|
Wang L, Yu Y, Guan H, Liu T, Qiao C. 67-kDa Laminin receptor contributes to hypoxia-induced migration and invasion of trophoblast-like cells by mediating matrix metalloproteinase-9. Clin Exp Pharmacol Physiol 2016; 42:549-58. [PMID: 25800042 DOI: 10.1111/1440-1681.12389] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/06/2015] [Accepted: 03/09/2015] [Indexed: 11/29/2022]
Abstract
Insufficient trophoblast invasion often occurs in patients experiencing preeclampsia. The 67-kDa laminin receptor (LR1) is a multifunctional protein that binds to laminin and interacts with the extracellular matrix. We recently demonstrated that LR1 is implicated in trophoblast migration and invasion. However, whether LR1 is involved in hypoxia-mediated trophoblastic invasion remains unclear and requires further investigation. This study demonstrates that two trophoblast-like cell lines (JEG3 and BeWo cells) cultured at 3% oxygen exerted enhanced migratory and invasive capabilities as compared with their counterparts exposed to 20% oxygen. LR1 expression was increased in hypoxic JEG3 cells but decreased after transfection with hypoxia-inducible factor 1 alpha (HIF-1α) specific siRNA. Moreover, shRNA targeting LR1 mRNA significantly inhibited hypoxia-induced increase in matrix metalloproteinase (MMP)-9 activity in JEG3 cells. Forced overexpression of LR1 augmented JEG3 cell migration and invasion, and enhanced MMP-9 expression and activity. Additionally, the blockade of the MMP-9 effect with its neutralizing antibody reduced LR1 elevation-promoted trophoblastic invasion. In summary, this study demonstrates that LR1 contributes to hypoxia-induced migration and invasion of trophoblast cells at least partly by mediating MMP-9 in vitro.
Collapse
Affiliation(s)
- Leilei Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | | | | | | | | |
Collapse
|
30
|
Roles of NGAL and MMP-9 in the tumor microenvironment and sensitivity to targeted therapy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:438-448. [PMID: 26278055 DOI: 10.1016/j.bbamcr.2015.08.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/08/2015] [Accepted: 08/10/2015] [Indexed: 12/15/2022]
Abstract
Various, diverse molecules contribute to the tumor microenvironment and influence invasion and metastasis. In this review, the roles of neutrophil gelatinase-associated lipocalin (NGAL) and matrix metalloproteinase-9 (MMP-9) in the tumor microenvironment and sensitivity to therapy will be discussed. The lipocalin family of proteins has many important functions. For example when NGAL forms a complex with MMP-9 it increases its stability which is important in cancer metastasis. Small hydrophobic molecules are bound by NGAL which can alter their entry into and efflux from cells. Iron transport and storage are also influenced by NGAL activity. Regulation of iron levels is important for survival in the tumor microenvironment as well as metastasis. Innate immunity is also regulated by NGAL as it can have bacteriostatic properties. NGAL and MMP-9 expression may also affect the sensitivity of cancer cells to chemotherapy as well as targeted therapy. Thus NGAL and MMP-9 play important roles in key processes involved in metastasis as well as response to therapy. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis, Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.
Collapse
|
31
|
Ebrahimi-Kalan A, Soleimani Rad J, Kafami L, Mohammadnejad D, Habibi Roudkenar M, Khaki AA, Aliyari Z, Mohammadi Roushandeh A. MS14 down-regulates lipocalin2 expression in spinal cord tissue in an animal model of multiple sclerosis in female C57BL/6. IRANIAN BIOMEDICAL JOURNAL 2015; 18:196-202. [PMID: 25326017 PMCID: PMC4225058 DOI: 10.6091/ibj.1375.2014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background: Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis, which is a demyelinating and an inflammatory disease of central nervous system. Recent studies have established that some molecules such as Lipocaline2 (LCN2), which expresses during inflammatory conditions, play an important role in EAE pathogenesis and might involve in its treatment process. Recently, it has been proved that MS14, an herbal-marine drug, has anti-inflammatory properties through reduction of TNF-α and IL-1β. Thus, the present study investigated the effects of MS14 on the course of EAE and its relation to LCN2 expression in both protein and gene levels. Methods: EAE was induced in female C57BL/6 mice using Hooke kits. Animals were scored for clinical signs of the disease according to a 10-point EAE scoring system. On 21st and 35th days after immunization, mice (n = 4/group) were deeply anesthetized, and the spinal cords were removed. Inflammatory cell infiltration and LCN2 expression in spinal cord were assessed by hematoxylin and eosin staining, immuno-histochemistry, and real-time PCR methods. Results: MS14 significantly ameliorated EAE symptoms and decreased lymphocyte infiltration into the spinal cord (P<0.05). Our data also revealed that LCN2 expression was significantly down-regulated in acute and chronic phases of EAE both at protein and gene levels after MS14 treatment (P<0.05). Conclusion: The results demonstrated that MS14 regulatory effect on EAE is accompanied by LCN2 down-regulation after treatment with the herb; however, more studies are required for clarifying the other involved mechanisms.
Collapse
Affiliation(s)
- Abbas Ebrahimi-Kalan
- Dept. of Neurosciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran;
- Shefa Neuroscience Research Center, Tehran, Iran;
| | - Jafar Soleimani Rad
- Dept. of Neurosciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran;
| | - Laya Kafami
- Shefa Neuroscience Research Center, Tehran, Iran;
- Dept. of Pathobiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran;
| | - Daryoush Mohammadnejad
- Dept. of Neurosciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran;
| | - Mehryar Habibi Roudkenar
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran;
| | - Amir Afshin Khaki
- Dept. of Neurosciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran;
| | - Zeynab Aliyari
- Dept. of Neurosciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran;
| | - Amaneh Mohammadi Roushandeh
- Dept. of Anatomical Sciences, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Corresponding Author; Mobile: (+98-914) 3078216; Fax: (+98-811) 838 0208; E-mail:
| |
Collapse
|