1
|
Tamas P, Farkas B, Betlehem J. Practical Considerations Concerning Preeclampsia Subgroups. J Clin Med 2025; 14:2498. [PMID: 40217948 PMCID: PMC11989745 DOI: 10.3390/jcm14072498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
Preeclampsia is one of the most serious clinical syndromes which can occur during pregnancy. According to our current knowledge, preeclampsia cannot be cured. However, a significant step forward is the recognizing preeclampsia is not a homogenous syndrome, i.e., different pathological events can lead to the hypertension + symptoms of organ damage, occurring in the second half of pregnancy. Clinically, two kinds of preeclampsia can be distinguished. The "classic" placental preeclampsia of immunological origin is characterized by contracted blood volume, fetal growth restriction, and marked alterations in laboratory indices. Patients in this subtype are characteristically young and primiparous. Clinical symptoms appear during the late second or early third trimester and show a quick progression. The outcome in cases of placental preeclampsia is frequently serious. For preventing the most critical conditions, the necessary delivery induction usually results in a preterm newborn. The maternal preeclampsia is associated with high blood volume. The characteristic augmented gestational weight gain is mostly a condition with a multifactorial background; however, obesity seems a critical risk factor. The early clinical symptoms are leg, and then generalized edema; hypertension and proteinuria appear after that. Laboratory abnormalities are rare; even platelet count remains within the normal range. The outcome is usually favorable; however, serious organ edema can lead to eclampsia or placental detachment. In the case of both types-from the name to the therapy-new data worthy of consideration have been created, which also justifies a change in attitude.
Collapse
Affiliation(s)
- Peter Tamas
- Doctoral School of Health Sciences, University of Pécs, H-7624 Pécs, Hungary (J.B.)
| | - Balint Farkas
- Department of Obstetrics and Gynecology, School of Medicine, University of Pécs, H-7624 Pécs, Hungary
| | - Jozsef Betlehem
- Doctoral School of Health Sciences, University of Pécs, H-7624 Pécs, Hungary (J.B.)
| |
Collapse
|
2
|
Williams BL, Zouache MA, Hageman GS. Author Response: Levels of the HtrA1 Protein in Serum and Vitreous Humor Are Independent of Genetic Risk for Age-Related Macular Degeneration at the 10q26 Locus. Invest Ophthalmol Vis Sci 2024; 65:26. [PMID: 39666349 PMCID: PMC11645747 DOI: 10.1167/iovs.65.14.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 12/13/2024] Open
Affiliation(s)
- Brandi L. Williams
- Sharon Eccles Steele Center for Translational Medicine, John A. Moran Eye Center, Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, Utah, United States. E-mail:
| | - Moussa A. Zouache
- Sharon Eccles Steele Center for Translational Medicine, John A. Moran Eye Center, Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, Utah, United States. E-mail:
| | - Gregory S. Hageman
- Sharon Eccles Steele Center for Translational Medicine, John A. Moran Eye Center, Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, Utah, United States. E-mail:
| |
Collapse
|
3
|
Degnes MHL, Westerberg AC, Andresen IJ, Henriksen T, Roland MCP, Zucknick M, Michelsen TM. Protein biomarker signatures of preeclampsia - a longitudinal 5000-multiplex proteomics study. Sci Rep 2024; 14:23654. [PMID: 39390022 PMCID: PMC11467422 DOI: 10.1038/s41598-024-73796-9] [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: 01/29/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
We aimed to explore novel biomarker candidates and biomarker signatures of late-onset preeclampsia (LOPE) by profiling samples collected in a longitudinal discovery cohort with a high-throughput proteomics platform. Using the Somalogic 5000-plex platform, we analyzed proteins in plasma samples collected at three visits (gestational weeks (GW) 12-19, 20-26 and 28-34 in 35 women with LOPE (birth ≥ 34 GW) and 70 healthy pregnant women). To identify biomarker signatures, we combined Elastic Net with Stability Selection for stable variable selection and validated their predictive performance in a validation cohort. The biomarker signature with the highest predictive performance (AUC 0.88 (95% CI 0.85-0.97)) was identified in the last trimester of pregnancy (GW 28-34) and included the Fatty acid amid hydrolase 2 (FAAH2), HtrA serine peptidase 1 (HTRA1) and Interleukin-17 receptor C (IL17RC) together with sFLT1 and maternal age, BMI and nulliparity. Our biomarker signature showed increased or similar predictive performance to the sFLT1/PGF-ratio within our data set, and we were able to validate the biomarker signature in a validation cohort (AUC ≥ 0.90). Further validation of these candidates should be performed using another protein quantification platform in an independent cohort where the negative and positive predictive values can be validly calculated.
Collapse
Affiliation(s)
- Maren-Helene Langeland Degnes
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway.
- Department of Biostatistics, Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway.
| | - Ane Cecilie Westerberg
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
| | - Ina Jungersen Andresen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
| | - Tore Henriksen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
| | - Marie Cecilie Paasche Roland
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
| | - Manuela Zucknick
- Department of Biostatistics, Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway
| | - Trond Melbye Michelsen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
4
|
Song S, Li X, Xue X, Dong W, Li C. Progress in the Study of the Role and Mechanism of HTRA1 in Diseases Related to Vascular Abnormalities. Int J Gen Med 2024; 17:1479-1491. [PMID: 38650587 PMCID: PMC11034561 DOI: 10.2147/ijgm.s456912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
High temperature requirement A1 (HTRA1) is a member of the serine protease family, comprising four structural domains: IGFBP domain, Kazal domain, protease domain and PDZ domain. HTRA1 encodes a serine protease, a secreted protein that is widely expressed in the vasculature. HTRA1 regulates a wide range of physiological processes through its proteolytic activity, and is also involved in a variety of vascular abnormalities-related diseases. This article reviews the role of HTRA1 in the development of vascular abnormalities-related hereditary cerebral small vessel disease (CSVD), age-related macular degeneration (AMD), tumors and other diseases. Through relevant research advances to understand the role of HTRA1 in regulating signaling pathways or refolding, translocation, degradation of extracellular matrix (ECM) proteins, thus directly or indirectly regulating angiogenesis, vascular remodeling, and playing an important role in vascular homeostasis, further understanding the mechanism of HTRA1's role in vascular abnormality-related diseases is important for HTRA1 to be used as a therapeutic target in related diseases.
Collapse
Affiliation(s)
- Shina Song
- Department of Neurology, The First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Department of Geriatrics, General Hospital of TISCO, Taiyuan, People’s Republic of China
| | - Xiaofeng Li
- Department of Neurology, The First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xuting Xue
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, The First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Wenping Dong
- Department of Geriatrics, General Hospital of TISCO, Taiyuan, People’s Republic of China
| | - Changxin Li
- Department of Neurology, The First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
5
|
Zhou W, Li X, Li X, Liu Y, Song W, Yang Q. The role of circular RNA in preeclampsia: From pathophysiological mechanism to clinical application. Life Sci 2024; 338:122407. [PMID: 38184270 DOI: 10.1016/j.lfs.2023.122407] [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: 11/05/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
Preeclampsia (PE) is a common pregnancy-induced hypertension disorder that poses a significant threat to the health of pregnant women and fetuses, and has become a leading cause of maternal, fetal, and neonatal mortality. Currently, the therapy strategy for PE is mainly prevention management and symptomatic treatment, and only delivery can completely terminate PE. Therefore, a deeper understanding of the pathogenesis of PE is needed to make treatment and prevention more effective and targeted. With the deepening of molecular etiology research, circular RNAs (circRNAs) have been found to be widely involved in various processes of PE pathogenesis. As a kind of RNA with a special "head to tail" loop structure, the characteristics of circRNAs enable them to play diverse roles in the pathophysiology of PE, and can also serve as ideal biomarkers for early prediction and monitoring progression of PE. In this review, we summarized the latest research on PE-related circRNAs, trying to elucidate the unique or shared roles of circRNAs in various pathophysiological mechanisms of PE, aiming to provide a whole picture of current research on PE-related circRNAs, and extend a new perspective for the precise screening and targeted therapy of PE.
Collapse
Affiliation(s)
- Wenjing Zhou
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China; Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiuying Li
- Medical Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.
| | - Xin Li
- Medical College, Jilin Engineering Vocational College, Siping, Jilin, China.
| | - Yaojia Liu
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China.
| | - Wenling Song
- Department of Obstetrics, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Qiwei Yang
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
6
|
Yoshida K, Kusama K, Shinohara G, Sato S, Yoshie M, Tamura K. Quercetin stimulates trophoblast fusion via the mitochondrial function. Sci Rep 2024; 14:287. [PMID: 38168580 PMCID: PMC10762005 DOI: 10.1038/s41598-023-50712-1] [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: 10/10/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
The fusion of mononuclear trophoblasts into multinucleate syncytiotrophoblasts is the critical event in the process of syncytialization, and its dysregulation can lead to pregnancy complications, notably hypertensive disorders of pregnancy (HDP). Oxidative stress may disrupt trophoblast syncytialization in HDP. Specifically, placentas with HDP exhibit impaired mitochondria, giving rise to the generation of reactive oxygen species (ROS) and subsequent oxidative stress. Quercetin, a bioflavonoid known for its antioxidant and anti-aging properties, has the potential to mitigate oxidative stress during trophoblast syncytialization. However, the precise mechanism underlying the action of quercetin in these processes remains to be elucidated. To explore the impact of quercetin on syncytialization, mitochondrial function, and ROS generation, cyclic AMP-stimulated BeWo cells were treated with quercetin. The expression of markers associated with cell fusion, mitochondrial function, and oxidative stress was determined using qPCR and western blotting. Additionally, morphological syncytialization and mitophagy (mitochondrial degradation) were assessed by immunofluorescence analysis. Our results revealed that quercetin increased the expression of syncytialization markers and promoted cell fusion. Furthermore, this compound also upregulated markers associated with mitophagy and mitochondrial fusion, which are corroborated by visual evidence of mitophagy through the fluorescence microscope. Cell fusion naturally stimulated ROS generation, which was attenuated by quercetin. Quercetin downregulated the expression of NRF2 and HO-1 during syncytialization, while increasing the expression of sirtuin1/3/6, which are known to play essential roles in antioxidant responses. In conclusion, quercetin effectively regulates mitochondrial function through its antioxidant properties and the suppression of ROS generation, ultimately promoting trophoblast fusion, suggesting that the flavonoid has the potential to ameliorate pregnancy-related disorder stemming from placental dysplasia.
Collapse
Affiliation(s)
- Kanoko Yoshida
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kazuya Kusama
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.
| | - Go Shinohara
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Shiho Sato
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Mikihiro Yoshie
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kazuhiro Tamura
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
7
|
Yuan X, Liu X, Zhu F, Huang B, Lin L, Huang J, Wen L, Kilby MD, Baker PN, Fu Y, Wu W, Qi H, Tang J, Tong C. Endoplasmic reticulum stress impairs trophoblast syncytialization through upregulation of HtrA4 and causes early-onset preeclampsia. J Hypertens 2023; 41:2095-2106. [PMID: 37728094 DOI: 10.1097/hjh.0000000000003541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
OBJECTIVE Syncytiotrophoblasts form via mononuclear cytotrophoblast fusion during placentation and play a critical role in maternal-fetal communication. Impaired syncytialization inevitably leads to pregnancy-associated complications, including preeclampsia. Endoplasmic reticulum stress (ERS) is reportedly linked with preeclampsia, but little is known about its association with syncytialization. High temperature requirement factor A4 (HtrA4), a placental-specific protease, is responsible for protein quality control and placental syncytialization. This study aimed to investigate the relationship among HtrA4, ERS, and trophoblast syncytialization in the development of early-onset preeclampsia (EO-PE). METHODS HtrA4 expression and ERS in preeclamptic placentas and control placentas were analyzed by Western blotting and qRT-PCR. HtrA4 and ERS localization in placentas was determined by immunohistochemistry and immunofluorescence. BeWo cells were used to stimulate the effects of HtrA4 and ERS on syncytialization. RESULTS HtrA4 expression was upregulated in EO-PE and positively correlated with ERS. HtrA4 activity was increased in preeclampsia. Under normoxia, HtrA4 overexpression in BeWo cells did not alter the ERS level. In addition, treatment with hypoxia/reoxygenation (H/R) or an ERS inducer increased HtrA4 expression. HtrA4 upregulation suppressed the levels of syncytin-2 and β-HCG in the presence of forskolin (FSK), and this change was exaggerated after ERS activation. In addition, treatment with an ERS inhibitor markedly suppressed FSK-treated cell fusion in a manner related to downregulation of HtrA4 expression. CONCLUSION Our results suggest that ERS enables syncytialization of placental development by upregulating HtrA4, but that excessive HtrA4 expression and preexisting ERS impair syncytialization and cause EO-PE.
Collapse
Affiliation(s)
- Xi Yuan
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Xiyao Liu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Fangyu Zhu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Biao Huang
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Li Lin
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Jiayu Huang
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | - Mark D Kilby
- Fetal Medicine Centre, Birmingham Women's & Children's Foundation Trust
- Institute of Metabolism & Systems Research, College of Medical & Dental Sciences, University of Birmingham, Birmingham
| | - Philip N Baker
- College of Life Sciences, University of Leicester, Leicester, UK
| | - Yong Fu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Weiwei Wu
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi
| | - Hongbo Qi
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
- Department of Obstetrics, Women and Children's Hospital of Chongqing Medical University
| | - Jing Tang
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
- School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Chao Tong
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| |
Collapse
|
8
|
Merle DA, Sen M, Armento A, Stanton CM, Thee EF, Meester-Smoor MA, Kaiser M, Clark SJ, Klaver CCW, Keane PA, Wright AF, Ehrmann M, Ueffing M. 10q26 - The enigma in age-related macular degeneration. Prog Retin Eye Res 2023; 96:101154. [PMID: 36513584 DOI: 10.1016/j.preteyeres.2022.101154] [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: 09/14/2022] [Revised: 11/21/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
Despite comprehensive research efforts over the last decades, the pathomechanisms of age-related macular degeneration (AMD) remain far from being understood. Large-scale genome wide association studies (GWAS) were able to provide a defined set of genetic aberrations which contribute to disease risk, with the strongest contributors mapping to distinct regions on chromosome 1 and 10. While the chromosome 1 locus comprises factors of the complement system with well-known functions, the role of the 10q26-locus in AMD-pathophysiology remains enigmatic. 10q26 harbors a cluster of three functional genes, namely PLEKHA1, ARMS2 and HTRA1, with most of the AMD-associated genetic variants mapping to the latter two genes. High linkage disequilibrium between ARMS2 and HTRA1 has kept association studies from reliably defining the risk-causing gene for long and only very recently the genetic risk region has been narrowed to ARMS2, suggesting that this is the true AMD gene at this locus. However, genetic associations alone do not suffice to prove causality and one or more of the 14 SNPs on this haplotype may be involved in long-range control of gene expression, leaving HTRA1 and PLEKHA1 still suspects in the pathogenic pathway. Both, ARMS2 and HTRA1 have been linked to extracellular matrix homeostasis, yet their exact molecular function as well as their role in AMD pathogenesis remains to be uncovered. The transcriptional regulation of the 10q26 locus adds an additional level of complexity, given, that gene-regulatory as well as epigenetic alterations may influence expression levels from 10q26 in diseased individuals. Here, we provide a comprehensive overview on the 10q26 locus and its three gene products on various levels of biological complexity and discuss current and future research strategies to shed light on one of the remaining enigmatic spots in the AMD landscape.
Collapse
Affiliation(s)
- David A Merle
- Institute for Ophthalmic Research, Department for Ophthalmology, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Department of Ophthalmology, Medical University of Graz, 8036, Graz, Austria.
| | - Merve Sen
- Institute for Ophthalmic Research, Department for Ophthalmology, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany
| | - Angela Armento
- Institute for Ophthalmic Research, Department for Ophthalmology, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany
| | - Chloe M Stanton
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Eric F Thee
- Department of Ophthalmology, Erasmus University Medical Center, 3015GD, Rotterdam, Netherlands; Department of Epidemiology, Erasmus University Medical Center, 3015CE, Rotterdam, Netherlands
| | - Magda A Meester-Smoor
- Department of Ophthalmology, Erasmus University Medical Center, 3015GD, Rotterdam, Netherlands; Department of Epidemiology, Erasmus University Medical Center, 3015CE, Rotterdam, Netherlands
| | - Markus Kaiser
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, 45117, Essen, Germany
| | - Simon J Clark
- Institute for Ophthalmic Research, Department for Ophthalmology, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus University Medical Center, 3015GD, Rotterdam, Netherlands; Department of Epidemiology, Erasmus University Medical Center, 3015CE, Rotterdam, Netherlands; Department of Ophthalmology, Radboudumc, 6525EX, Nijmegen, Netherlands; Institute of Molecular and Clinical Ophthalmology Basel, CH-4031, Basel, Switzerland
| | - Pearse A Keane
- Institute for Health Research, Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital NHS Foundation Trust, UCL Institute of Ophthalmology, London, EC1V 2PD, UK
| | - Alan F Wright
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Michael Ehrmann
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, 45117, Essen, Germany
| | - Marius Ueffing
- Institute for Ophthalmic Research, Department for Ophthalmology, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany.
| |
Collapse
|
9
|
Wu Y, Liu Y, Ding Y. Predictive Performance of Placental Protein 13 for Screening Preeclampsia in the First Trimester: A Systematic Review and Meta-Analysis. Front Med (Lausanne) 2021; 8:756383. [PMID: 34869456 PMCID: PMC8640131 DOI: 10.3389/fmed.2021.756383] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/25/2021] [Indexed: 12/02/2022] Open
Abstract
Preeclampsia is a pregnancy-specific syndrome that affects maternal and neonatal mortality. Several serum biomarkers can be used to predict preeclampsia. Among these proteins, placental protein 13 (PP13) has received progressively more interest in recent studies. The decrease in PP13 expression is one of the earliest signs for the development of preeclampsia and has shown its predictive performance for preeclampsia. In this meta-analysis, we collected 17 observational studies with 40,474 pregnant women. The overall sensitivity of PP13 to predict preeclampsia was 0.62 [95% confidence interval (CI) = 0.49–0.74], the specificity was 0.84 (95%CI = 0.81–0.86), and the diagnostic odds ratio was nine (95%CI = 5–15). The area under the curve for summary receiver operating characteristic was 0.84. We then chose the early-onset preeclampsia as a subgroup. The sensitivity of early-onset subgroup was 0.63 (95%CI = 0.58–0.76), the specificity was 0.85 (95%CI = 0.82–0.88), and the diagnostic odds ratio was 10 (95%CI = 6–18). The findings of our meta-analysis indicate that PP13 may be an effective serum biomarker for the predictive screening of preeclampsia. Nonetheless, large prospective cohort studies and randomized controlled trials are expected to uncover its application in clinical practice. The heterogeneity of the original trials may limit the clinical application of PP13. Systematic Review Registration:https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=188948 The meta-analysis was registered in PROSPERO (CRD42020188948).
Collapse
Affiliation(s)
- Yifan Wu
- Department of Obstetrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Obstetrics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Liu
- Department of Obstetrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yiling Ding
- Department of Obstetrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Overview of Human HtrA Family Proteases and Their Distinctive Physiological Roles and Unique Involvement in Diseases, Especially Cancer and Pregnancy Complications. Int J Mol Sci 2021; 22:ijms221910756. [PMID: 34639128 PMCID: PMC8509474 DOI: 10.3390/ijms221910756] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/27/2021] [Accepted: 10/02/2021] [Indexed: 12/25/2022] Open
Abstract
The mammalian high temperature requirement A (HtrA) proteins are a family of evolutionarily conserved serine proteases, consisting of four homologs (HtrA1-4) that are involved in many cellular processes such as growth, unfolded protein stress response and programmed cell death. In humans, while HtrA1, 2 and 3 are widely expressed in multiple tissues with variable levels, HtrA4 expression is largely restricted to the placenta with the protein released into maternal circulation during pregnancy. This limited expression sets HtrA4 apart from the rest of the family. All four HtrAs are active proteases, and their specific cellular and physiological roles depend on tissue type. The dysregulation of HtrAs has been implicated in many human diseases such as cancer, arthritis, neurogenerative ailments and reproductive disorders. This review first discusses HtrAs broadly and then focuses on the current knowledge of key molecular characteristics of individual human HtrAs, their similarities and differences and their reported physiological functions. HtrAs in other species are also briefly mentioned in the context of understanding the human HtrAs. It then reviews the distinctive involvement of each HtrA in various human diseases, especially cancer and pregnancy complications. It is noteworthy that HtrA4 expression has not yet been reported in any primary tumour samples, suggesting an unlikely involvement of this HtrA in cancer. Collectively, we accentuate that a better understanding of tissue-specific regulation and distinctive physiological and pathological roles of each HtrA will improve our knowledge of many processes that are critical for human health.
Collapse
|
11
|
Campbell RA, Campbell HD, Bircher JS, de Araujo CV, Denorme F, Crandell JL, Rustad JL, Monts J, Cody MJ, Kosaka Y, Yost CC. Placental HTRA1 cleaves α1-antitrypsin to generate a NET-inhibitory peptide. Blood 2021; 138:977-988. [PMID: 34192300 PMCID: PMC9069473 DOI: 10.1182/blood.2020009021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 06/30/2021] [Accepted: 06/13/2021] [Indexed: 11/20/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are important components of innate immunity. Neonatal neutrophils (polymorphonuclear leukocytes [PMNs]) fail to form NETs due to circulating NET-inhibitory peptides (NIPs), cleavage fragments of α1-antitrypsin (A1AT). How fetal and neonatal blood NIPs are generated remains unknown, however. The placenta expresses high-temperature requirement serine protease A1 (HTRA1) during fetal development, which can cleave A1AT. We hypothesized that placentally expressed HTRA1 regulates the formation of NIPs and that NET competency changed in PMNs isolated from neonatal HTRA1 knockout mice (HTRA1-/-). We found that umbilical cord blood plasma has elevated HTRA1 levels compared with adult plasma and that recombinant and placenta-eluted HTRA1 cleaves A1AT to generate an A1AT cleavage fragment (A1ATM383S-CF) of molecular weight similar to previously identified NIPs that block NET formation by adult neutrophils. We showed that neonatal mouse pup plasma contains A1AT fragments that inhibit NET formation by PMNs isolated from adult mice, indicating that NIP generation during gestation is conserved across species. Lipopolysaccharide-stimulated PMNs isolated from HTRA1+/+ littermate control pups exhibit delayed NET formation after birth. However, plasma from HTRA1-/- pups had no detectable NIPs, and PMNs from HTRA1-/- pups became NET competent earlier after birth compared with HTRA1+/+ littermate controls. Finally, in the cecal slurry model of neonatal sepsis, A1ATM383S-CF improved survival in C57BL/6 pups by preventing pathogenic NET formation. Our data indicate that placentally expressed HTRA1 is a serine protease that cleaves A1AT in utero to generate NIPs that regulate NET formation by human and mouse PMNs.
Collapse
Affiliation(s)
- Robert A Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
- Department of Internal Medicine
| | | | | | | | - Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - Jacob L Crandell
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - John L Rustad
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - Josh Monts
- Flow Cytometry Core, University of Utah, Salt Lake City, UT
| | - Mark J Cody
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
- Department of Pediatrics, and
| | - Yasuhiro Kosaka
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - Christian C Yost
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
- Department of Pediatrics, and
| |
Collapse
|
12
|
Hjæresen S, Sejbaek T, Axelsson M, Vinsløv-Jensen H, Mortensen SK, Pihl-Jensen G, Novakova L, Christensen JDR, Pedersen CB, Halle B, Poulsen FR, Lautrup Frederiksen J, Zhang M, Benedikz E, Lycke J, Illes Z, Fex Svenningsen Å. The levels of the serine protease HTRA1 in cerebrospinal fluid correlate with progression and disability in multiple sclerosis. J Neurol 2021; 268:3316-3324. [PMID: 33661357 DOI: 10.1007/s00415-021-10489-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND High Temperature Requirement Serine Protease A1 (HTRA1) degrades extracellular matrix molecules (ECMs) and growth factors. It interacts with several proteins implicated in multiple sclerosis (MS), but has not previously been linked to the disease. OBJECTIVE Investigate the levels of HTRA1 in cerebrospinal fluid (CSF) in different subtypes of MS and brain tissue. METHODS Using ELISA, HTRA1 levels were compared in CSF from untreated patients with relapsing-remitting MS (RRMS, n = 23), secondary progressive MS (SPMS, n = 26) and healthy controls (HCs, n = 26). The effect of disease modifying therapies (DMTs) were examined in both patient groups. Cellular distribution in human brain was studied using immunochemistry and the oligointernode database, based on a single-nuclei RNA expression map. RESULTS HTRA1 increased in RRMS and SPMS compared to HCs. DMT decreased HTRA1 levels in both types of MS. Using ROC analysis, HTRA1 cut-offs could discriminate HCs from RRMS patients with 100% specificity and 82.6% sensitivity. In the brain, HTRA1 was expressed in glia and neurons. CONCLUSION HTRA1 is a promising CSF biomarker for MS correlating with disease- and disability progression. Most cell species of the normal and diseased CNS express HTRA1 and the expression pattern could reflect pathological processes involved in MS pathogenesis.
Collapse
Affiliation(s)
- Simone Hjæresen
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Tobias Sejbaek
- Department of Regional Health Research, University of Southern Denmark, 5000, Odense, Denmark
- Department of Neurology, Southwest Jutland University Hospital, 6700, Esbjerg, Denmark
| | - Marcus Axelsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, and Department of Neurology, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Helle Vinsløv-Jensen
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
| | - Sif Kløvedal Mortensen
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
| | - Gorm Pihl-Jensen
- Department of Neurology, University of Copenhagen, Rigshospitalet Glostrup, Copenhagen, Denmark
| | - Lenka Novakova
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, and Department of Neurology, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Christian Bonde Pedersen
- Department of Neurosurgery, Clinical Institute, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Bo Halle
- Department of Neurosurgery, Clinical Institute, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Frantz Rom Poulsen
- Department of Neurosurgery, Clinical Institute, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | | | - Mengliang Zhang
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Eirikur Benedikz
- Faculty of Health Sciences, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
| | - Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, and Department of Neurology, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Zsolt Illes
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
- Department of Neurology, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Åsa Fex Svenningsen
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark.
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
13
|
Fantone S, Giannubilo SR, Marzioni D, Tossetta G. HTRA family proteins in pregnancy outcome. Tissue Cell 2021; 72:101549. [PMID: 33915357 DOI: 10.1016/j.tice.2021.101549] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/02/2021] [Accepted: 04/17/2021] [Indexed: 11/29/2022]
Abstract
HTRA (High temperature requirement protease A) family proteins includes HTRA1 (L56 or PRSS11), HTRA2/Omi, HTRA3 (PRSP) and HTRA4. These are oligomeric serine proteases highly conserved from bacteria to humans and are involved in a variety of biological functions including the maintenance of normal cell physiology and pathogenicity such as cell growth, apoptosis, neurodegenerative disorders, inflammation diseases and cancer. These proteins are normally expressed in placental villi during all pregnancy but their expression is found to be altered in pathological pregnancies suggesting a possible role of those proteins in the development of human placenta. Moreover, some HTRA family proteins have also been found in maternal blood and were impaired in pathological pregnancy suggesting a possible role of some of these proteins as early markers of pregnancy outcome. The aim of this review is to summarize the data currently available on the role of HTRA family proteins in pregnancy focalizing their role in pregnancy complications such as Preeclampsia (PE), IntraUterine Growth Restriction (IUGR) and Spontaneus PreTerm Birth (SPTB).
Collapse
Affiliation(s)
- Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Stefano R Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, Ancona, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy; Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, Ancona, Italy.
| |
Collapse
|
14
|
Skinner S, Rolnik DL, Wang Y, Nie G, Syngelaki A, Nicolaides KH, da Silva Costa F. Second and third trimester serum levels of HtrA1 in pregnancies affected by pre-eclampsia. Placenta 2021; 106:1-6. [PMID: 33601219 DOI: 10.1016/j.placenta.2021.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/21/2020] [Accepted: 02/01/2021] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Altered placental expression of high temperature requirement factor A1 (HtrA1) is implicated in abnormal trophoblastic invasion and endothelial dysfunction in pre-eclampsia (PE). Serum levels of HtrA1 have been proposed as a novel biomarker to improve the prediction of PE. This study assesses serum HtrA1 levels in prospectively collected samples of women who developed PE compared to normotensive pregnancies. METHODS This was a case-control study of serum HtrA1 levels in second and third trimester samples in women who later developed preterm or term PE compared to controls. Overall, 300 serum samples were drawn from a prospective observational study of adverse pregnancy outcomes in three different gestational age windows (19-24, 30-34 and 35-37 weeks) at the Fetal Medicine Research Institute, King's College Hospital, London. Serum HtrA1 levels were determined by enzyme-linked immunosorbent assay (ELISA) by a blinded laboratory professional. Median HtrA1 MoM values, adjusted for gestational age and maternal characteristics, were compared between cases and controls at each gestational age group. RESULTS Women who later developed PE, compared to controls, had significantly higher maternal weight and more frequently had chronic hypertension or a history of PE in a previous pregnancy. In normotensive pregnancies, serum HtrA1 increased with increasing gestational age, whereas, in PE pregnancies HtrA1 levels remained stable, but were not significantly different from control pregnancies at any gestational age. DISCUSSION Serum HtrA1 levels are not significantly different in women who develop PE compared to controls.
Collapse
Affiliation(s)
- Sasha Skinner
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia.
| | - Daniel L Rolnik
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Yao Wang
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Guiying Nie
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia; School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Argyro Syngelaki
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | | | - Fabricio da Silva Costa
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia; Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
15
|
Owen LA, Shirer K, Collazo SA, Szczotka K, Baker S, Wood B, Carroll L, Haaland B, Iwata T, Katikaneni LD, DeAngelis MM. The Serine Protease HTRA-1 Is a Biomarker for ROP and Mediates Retinal Neovascularization. Front Mol Neurosci 2020; 13:605918. [PMID: 33281553 PMCID: PMC7705345 DOI: 10.3389/fnmol.2020.605918] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 10/22/2020] [Indexed: 01/29/2023] Open
Abstract
Retinopathy of prematurity (ROP) is a blinding aberrancy of retinal vascular maturation in preterm infants. Despite delayed onset after preterm birth, representing a window for therapeutic intervention, we cannot prevent or cure ROP blindness. A natural form of ROP protection exists in the setting of early-onset maternal preeclampsia, though is not well characterized. As ischemia is a central feature in both ROP and preeclampsia, we hypothesized that angiogenesis mediators may underlie this protection. To test our hypothesis we analyzed peripheral blood expression of candidate proteins with suggested roles in preeclamptic and ROP pathophysiology and with a proposed angiogenesis function (HTRA-1, IGF-1, TGFβ-1, and VEGF-A). Analysis in a discovery cohort of 40 maternal-infant pairs found that elevated HTRA-1 (high-temperature requirement-A serine peptidase-1) was significantly associated with increased risk of ROP and the absence of preeclampsia, thus fitting a model of preeclampsia-mediated ROP protection. We validated these findings and further demonstrated a dose-response between systemic infant HTRA-1 expression and risk for ROP development in a larger and more diverse validation cohort consisting of preterm infants recruited from two institutions. Functional analysis in the oxygen-induced retinopathy (OIR) murine model of ROP supported our systemic human findings at the local tissue level, demonstrating that HtrA-1 expression is elevated in both the neurosensory retina and retinal pigment epithelium by RT-PCR in the ROP disease state. Finally, transgenic mice over-expressing HtrA-1 demonstrate greater ROP disease severity in this model. Thus, HTRA-1 may underlie ROP protection in preeclampsia and represent an avenue for disease prevention, which does not currently exist.
Collapse
Affiliation(s)
- Leah A Owen
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, United States
| | - Kinsey Shirer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, United States
| | - Samuel A Collazo
- School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Kathryn Szczotka
- Department of Obstetrics and Gynecology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Shawna Baker
- Center for Clinical and Translational Science, University of Utah, Salt Lake City, UT, United States
| | - Blair Wood
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, United States
| | - Lara Carroll
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, United States
| | - Benjamin Haaland
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, United States
| | - Takeshi Iwata
- National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Lakshmi D Katikaneni
- Department of Pediatrics, Division of Neonatology, Medical University of South Carolina, Charleston, SC, United States
| | - Margaret M DeAngelis
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, United States.,Department of Pharmacotherapy, The College of Pharmacy, University of Utah, Salt Lake City, UT, United States.,Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences State University of New York, Buffalo, NY, United States
| |
Collapse
|
16
|
Ou Y, Zhu L, Wei X, Bai S, Chen M, Chen H, Zhang J. Circular RNA circ_0111277 attenuates human trophoblast cell invasion and migration by regulating miR-494/HTRA1/Notch-1 signal pathway in pre-eclampsia. Cell Death Dis 2020; 11:479. [PMID: 32587240 PMCID: PMC7316814 DOI: 10.1038/s41419-020-2679-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 05/23/2020] [Accepted: 05/26/2020] [Indexed: 11/25/2022]
Abstract
Mounting evidence has revealed that impaired spiral artery remodeling, placental dysfunction, and inadequate trophoblast invasion are closely correlated with the etiology and pathogenesis of pre-eclampsia (PE). Moreover, defective trophoblast invasion may trigger poor maternal-fetal circulation and placental hypoxia, leading to PE. However, the detailed molecular pathology of PE remains unclear. Although circRNAs, as a new type of stable and abundant endogenous noncoding RNA, have been proven to be essential to the pathogenesis of various diseases, their role in PE requires further verification. In this context, it is necessary to unveil the roles of circRNAs in regulating the migration and invasion of extravillous trophoblasts. In this study, using quantitative real-time PCR, we confirmed that hsa_circ_0111277 was upregulated in PE placentas relative to the level in normal pregnancy placentas. In addition, positive correlations between hsa_circ_0111277 expression and PE-related factors (proteinuria level at 24 h and placental weight) were identified by Pearson's analysis based on the clinical data of 25 PE patients. Moreover, fluorescence in situ hybridization analysis illustrated that circ_0111277 was preferentially localized within the cytoplasm. Mechanistically, circ_0111277 sponged hsa-miR-494-3p in trophoblast cells to attenuate the latter's repression by regulating HTRA1/Notch-1 expression. In conclusion, trophoblast cell migration and invasion were shown to be promoted and modulated by the hsa_circ_0111277/miR-494-3p/HTRA1/Notch-1 axis, which provides useful insight for exploring a new therapeutic approach for PE.
Collapse
Affiliation(s)
- Yuhua Ou
- Department of Obstetrics and Gynecology, Guangdong Women and Children Hospital, Guangzhou, 511400, Guangdong, China
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Liqiong Zhu
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Xiangcai Wei
- Department of Obstetrics and Gynecology, Guangdong Women and Children Hospital, Guangzhou, 511400, Guangdong, China
| | - Shiyu Bai
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Manqi Chen
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Hui Chen
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China.
| | - Jianping Zhang
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
17
|
Carroll L, Owen LA. Current evidence and outcomes for retinopathy of prematurity prevention: insight into novel maternal and placental contributions. EXPLORATION OF MEDICINE 2020; 1:4-26. [PMID: 32342063 PMCID: PMC7185238 DOI: 10.37349/emed.2020.00002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a blinding morbidity of preterm infants, which represents a significant clinical problem, accounting for up to 40% of all childhood blindness. ROP displays a range of severity, though even mild disease may result in life-long visual impairment. This is complicated by the fact that our current treatments have significant ocular and potentially systemic effects. Therefore, disease prevention is desperately needed to mitigate the life-long deleterious effects of ROP for preterm infants. Although ROP demonstrates a delayed onset of retinal disease following preterm birth, representing a potential window for prevention, we have been unable to sufficiently alter the natural disease course and meaningfully prevent ROP. Prevention therapeutics requires knowledge of early ROP molecular changes and risk, occurring prior to clinical retinal disease. While we still have an incomplete understanding of these disease mechanisms, emerging data integrating contributions of maternal/placental pathobiology with ROP are poised to inform novel approaches to prevention. Herein, we review the molecular basis for current prevention strategies and the clinical outcomes of these interventions. We also discuss how insights into early ROP pathophysiology may be gained by a better understanding of maternal and placental factors playing a role in preterm birth.
Collapse
Affiliation(s)
- Lara Carroll
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 4132, USA
| | - Leah A. Owen
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 4132, USA
| |
Collapse
|
18
|
Supanji S, Bahana Ilham Perdamaian A, Dianratri A, Syifarahmah A, Wahyu Widayanti T, Setya Wardhana F, Bayu Sasongko M, Eko Prayogo M, Nurini Agni A, Oka C. HtrA1 serine protease expression levels on age-related macular degeneration (AMD) patients in Yogyakarta. BIO WEB OF CONFERENCES 2020. [DOI: 10.1051/bioconf/20202802004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This research aims to investigate the HtrA1 serine protease circulating level of Age-related Macular Degeneration (AMD) patients in Yogyakarta, Indonesia. This study was conducted from January to August 2019 which included 38 AMD patients and 16 Non-AMD patients/controls (two groups). Baseline data and blood sample were collected. ELISA assay was used to measure the HtrA1 serine protease circulating level on both groups. SNP genotyping of rs10490924 was using restriction enzyme digestion. This study used The IBM SPSS® version 24 (Chicago, The USA) to determine the relationship between HtrA1 expression level and AMD incidence. AMD patients had higher HtrA1 serine protease level (35.31) than controls (30.08). However, there is no association found between HtrA1 serine protease level and AMD incidence (p-value>0.05, CI 95 %). However, HtrA1 serine protease did not associate positively to AMD incidence in Yogyakarta samples. Further analysis by grouping AMD patient based on the rs10490924 genotype show no statistical correlation between HTRA1 to the incidence of AMD. This result might be due to the lack of samples in the study groups. Future studies with larger number of samples are advised to better see the association between Htra1 serine protease level and AMD incidence.
Collapse
|
19
|
Abstract
Pre-eclampsia is a common disorder that particularly affects first pregnancies. The clinical presentation is highly variable but hypertension and proteinuria are usually seen. These systemic signs arise from soluble factors released from the placenta as a result of a response to stress of syncytiotrophoblast. There are two sub-types: early and late onset pre-eclampsia, with others almost certainly yet to be identified. Early onset pre-eclampsia arises owing to defective placentation, whilst late onset pre-eclampsia may center around interactions between normal senescence of the placenta and a maternal genetic predisposition to cardiovascular and metabolic disease. The causes, placental and maternal, vary among individuals. Recent research has focused on placental-uterine interactions in early pregnancy. The aim now is to translate these findings into new ways to predict, prevent, and treat pre-eclampsia.
Collapse
Affiliation(s)
- Graham J Burton
- Department of Physiology, Development & Neuroscience, University of Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, UK
| | | | - James M Roberts
- Magee-Womens Research Institute, Depts. Obstetric Gynecology and Reproductive Sciences, Epidemiology, and Clinical and Translational Research, University of Pittsburgh, USA
| | - Ashley Moffett
- Centre for Trophoblast Research, University of Cambridge, UK
- Dept of Pathology, University of Cambridge, UK
| |
Collapse
|
20
|
Teoh SSY, Wang Y, Li Y, Leemaqz SYL, Dekker GA, Roberts CT, Nie G. Low Serum Levels of HtrA3 at 15 Weeks of Gestation Are Associated with Late-Onset Preeclampsia Development and Small for Gestational Age Birth. Fetal Diagn Ther 2019; 46:392-401. [PMID: 31013509 DOI: 10.1159/000497144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/21/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the potential utility of serum HtrA1 and HtrA3, serine proteases that are highly expressed in the developing placenta, at 15 and 20 weeks of gestation for predicting later development of adverse pregnancy outcomes of preeclampsia (PE), gestational hypertension (GHT), preterm birth (PTB), and small for gestational age (SGA) birth. METHODS This is a nested case control study of 665 samples (330 controls, 335 cases) from the Adelaide SCOPE cohort. The cases included were 92 PE, 71 GHT, 56 PTB, and 116 SGA. Samples were assessed by ELISA and data adjusted for maternal age, BMI, socioeconomic index, hCG, and smoking status. Multivariate logistic regression was performed with other biochemical and biophysical parameters available for these samples. RESULTS HtrA1 did not differ between the controls and cases. In contrast, HtrA3 was significantly lower at 15 weeks in pregnancies that later developed late-onset PE (LPE) or resulted in SGA birth, with an area under the ROC curve (AUC) of 0.716 and 0.790, respectively. The combination of HtrA3 with PAPP-A, uterine, and umbilical Doppler improved the AUC to 0.755 for LPE and 0.844 for SGA. CONCLUSION HtrA3 at 15 weeks is associated with, and may be useful for, the early detection of LPE development and SGA birth.
Collapse
Affiliation(s)
- Sonia Soo Yee Teoh
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Yao Wang
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Ying Li
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Shalem Yiner-Lee Leemaqz
- Robinson Research Institute and School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Gus A Dekker
- Robinson Research Institute and School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Claire T Roberts
- Robinson Research Institute and School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Guiying Nie
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia,
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia,
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia,
| |
Collapse
|
21
|
Wang Y, Lim R, Nie G. HtrA4 may play a major role in inhibiting endothelial repair in pregnancy complication preeclampsia. Sci Rep 2019; 9:2728. [PMID: 30804477 PMCID: PMC6389976 DOI: 10.1038/s41598-019-39565-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/15/2019] [Indexed: 12/03/2022] Open
Abstract
Preeclampsia (PE) is a life-threatening complication of human pregnancy with no effective treatment other than premature delivery. It is hallmarked by systemic endothelial injury/dysfunction which is believed to be caused by abnormal levels/types of placenta-derived factors that are circulating in the maternal blood. Emerging evidence suggests that endothelial repair is also dysregulated in PE, as circulating endothelial progenitor cells (EPCs) critical for endothelial regeneration are reduced in number and functionality. However, the underlying mechanisms are poorly understood. HtrA4 is a placenta-specific protease that is secreted into the circulation and significantly elevated in early-onset PE. Here we investigated the impact of HtrA4 on endothelial proliferation and repair. We demonstrated that high levels of HtrA4 halted endothelial cell proliferation and significantly down-regulated a number of genes that are critical for cell cycle progression, including CDKN3, BIRC5, CDK1 and MKI67. Furthermore, HtrA4 significantly inhibited the proliferation of primary EPCs isolated from term human umbilical cord blood and impeded their differentiation into mature endothelial cells. Our data thus suggests that elevated levels of HtrA4 in the early-onset PE circulation may impair endothelial cell repair, not only by halting endothelial cell proliferation, but also by inhibiting the proliferation and differentiation of circulating EPCs.
Collapse
Affiliation(s)
- Yao Wang
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3800, Australia
| | - Rebecca Lim
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, 3168, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, 3168, Australia
| | - Guiying Nie
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia. .,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3800, Australia. .,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
22
|
Cheng H, Zhu H, Cao M, Lu C, Bao S, Pan Y. HtrA1 suppresses the growth of pancreatic cancer cells by modulating Notch-1 expression. Braz J Med Biol Res 2018; 52:e7718. [PMID: 30484491 PMCID: PMC6262754 DOI: 10.1590/1414-431x20187718] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/05/2018] [Indexed: 01/09/2023] Open
Abstract
Pancreatic cancer is well known to be the most deadly malignancy with the worst survival rate of all cancers. High temperature requirement factor A1 (HtrA1) plays an important role in cancer cell proliferation, migration, apoptosis, and differentiation. This study aimed to explore the function of HtrA1 in pancreatic cancer cell growth and its underlying mechanism. We found that the expression of HtrA1 was lower in pancreatic cancer tissue compared to the adjacent normal tissue. Consistently, HtrA1 levels were also decreased in two human pancreatic cancer cell lines, PANC-1 and BXPC-3. Moreover, enforced expression of HtrA1 inhibited cell viability and colony formation of PANC-1 and BXPC-3 cells. Overexpression of HtrA1 promoted apoptosis and suppressed migratory ability of tumor cells. On the contrary, siRNA-mediated knockdown of HtrA1 promoted the growth potential of pancreatic cancer cells. In addition, we found that up-regulation of HtrA1 reduced the expression of Notch-1 in pancreatic cancer cells. On the contrary, knockdown of HtrA1 increased the expression levels of Notch-1. Furthermore, overexpression of Notch-1 abolished the anti-proliferative effect of HtrA1 on pancreatic cancer cells. Taken together, our findings demonstrated that HtrA1 could inhibit pancreatic cancer cell growth via regulating Notch-1 expression, which implied that HtrA1 might be developed as a novel molecular target for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Hao Cheng
- Department of General Surgery, The Afflicted Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Hao Zhu
- Department of Gastroenterology, The Afflicted Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Meng Cao
- Department of General Surgery, The Afflicted Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Chenglin Lu
- Department of General Surgery, The Afflicted Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Shanhua Bao
- Department of General Surgery, The Afflicted Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yiming Pan
- Department of General Surgery, The Afflicted Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
23
|
Klose R, Adam MG, Weis EM, Moll I, Wüstehube-Lausch J, Tetzlaff F, Oka C, Ehrmann M, Fischer A. Inactivation of the serine protease HTRA1 inhibits tumor growth by deregulating angiogenesis. Oncogene 2018; 37:4260-4272. [PMID: 29713059 DOI: 10.1038/s41388-018-0258-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 02/26/2018] [Accepted: 03/27/2018] [Indexed: 11/09/2022]
Abstract
The serine protease HTRA1 is involved in several vascular diseases and its expression is often deregulated in cancer. We aimed at identifying how HTRA1 in the vasculature affects tumor growth. Here we report that silencing of HTRA1 in cultured endothelial cells increased migration rate and tube formation, whereas forced HTRA1 expression impaired sprouting angiogenesis. Mechanistically, endothelial HTRA1 expression enhanced Delta/Notch signaling by reducing the amount of the weak Notch ligand JAG1. HTRA1 physically interacted with JAG1 and cleaved it within the intracellular domain, leading to protein degradation. Expression of a constitutive active Notch1 prevented the hypersprouting phenotype upon silencing of HTRA1. In HtrA1-deficient mice, endothelial Notch signaling was diminished and isolated endothelial cells had increased expression of VEGF receptor-2. Growth of syngeneic tumors was strongly impaired in HtrA1-/- mice. The tumor vasculature was much denser in HtrA1-/- mice and less covered with mural cells. This chaotic and immature vascular network was poorly functional as indicated by large hypoxic tumor areas and low tumor cell proliferation rates. In summary, inhibition of HTRA1 in the tumor stroma impaired tumor progression by deregulating angiogenesis.
Collapse
Affiliation(s)
- Ralph Klose
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - M Gordian Adam
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany.,Metanomics Health GmbH, Berlin, Germany
| | - Eva-Maria Weis
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - Joycelyn Wüstehube-Lausch
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.,BioNTech AG, Mainz, Germany
| | - Fabian Tetzlaff
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - Chio Oka
- Laboratory of Gene Function in Animals, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara, 630-0192, Japan
| | - Michael Ehrmann
- Centre of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, 45117, Germany
| | - Andreas Fischer
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany. .,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany. .,Medical Clinic I, Endocrinology and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, 69120, Germany.
| |
Collapse
|
24
|
Xiang H, Tao X, Xia S, Qu J, Song H, Liu J, Shang D. Emodin Alleviates Sodium Taurocholate-Induced Pancreatic Acinar Cell Injury via MicroRNA-30a-5p-Mediated Inhibition of High-Temperature Requirement A/Transforming Growth Factor Beta 1 Inflammatory Signaling. Front Immunol 2017; 8:1488. [PMID: 29163548 PMCID: PMC5681496 DOI: 10.3389/fimmu.2017.01488] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022] Open
Abstract
Pancreatitis is an inflammatory disease that is responsible for substantial morbidity and mortality, and it can induce pancreatic necrosis that starts within pancreatic acinar cells in severe cases. Emodin, a pleiotropic natural product isolated from the Chinese herb Rheum palmatum L., has effective anti-inflammatory activities. In this paper, we investigated the protective effects and molecular mechanism of emodin against sodium taurocholate (STC)-induced pancreatic acinar cells injury in vitro and in vivo; and the results showed that emodin could significantly alleviate STC-induced pancreatic acinar cells injury through decreasing trypsin, amylase and the release of inflammatory factors (tumor necrosis factor alpha, interleukin-1β, and interleukin-6). Also, we found that emodin could significantly downregulate the HTRA1, interleukin-33, myeloid differentiation primary response gene 88, TNF receptor-associated factor-6, and nuclear factor kappa-B protein levels, but upregulate the transforming growth factor beta 1 (TGF-β1) protein level. These results indicated that emodin alleviated pancreatic acinar cells injury mainly through inhibiting HTRA1/TGF-β1 signaling pathway, and this finding was further proved by the HTRA1 overexpression experiments. In addition, the inflammatory regulator microRNA-30a-5p (miR-30a-5p) was confirmed to be a transcriptional brake that controls the HTRA1 gene through using a dual luciferase reporter assay, and it was upregulated by emodin in pancreatic acinar cells. Furthermore, the pancreatic protective effects and anti-inflammatory activities of emodin were all abrogated with both miR-30a-5p inhibitor in vitro and miR-30a-5p antagomir in vivo. Collectively, these results demonstrate that miR-30a-5p/HTRA1 are the target of emodin-mediated attenuation of pancreatic acinar cell injury in pancreatitis, thus providing the foundation for further development of this natural product for medical therapy.
Collapse
Affiliation(s)
- Hong Xiang
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xufeng Tao
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Shilin Xia
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jialin Qu
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huiyi Song
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jianjun Liu
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
25
|
Wang Y, Chen Q, Zhao M, Walton K, Harrison C, Nie G. Multiple Soluble TGF-β Receptors in Addition to Soluble Endoglin Are Elevated in Preeclamptic Serum and They Synergistically Inhibit TGF-β Signaling. J Clin Endocrinol Metab 2017; 102. [PMID: 28633389 PMCID: PMC5546862 DOI: 10.1210/jc.2017-01150] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Preeclampsia (PE) can be classified into early-onset (<34 weeks of gestation) and late-onset (>34 weeks of gestation) subtypes. Soluble endoglin, an auxiliary receptor for transforming growth factor (TGF)-β ligands, is increased in PE circulation and believed to inhibit TGF-β action by sequestering the ligands. However, soluble endoglin, with a low affinity to TGF-β ligands, has been demonstrated to have little effect by itself on TGF-β action. OBJECTIVES We examined whether multiple soluble TGF-β receptors are elevated in PE circulation and whether they synergistically block TGF-β signaling. DESIGN TGF-β receptors were measured using enzyme-linked immunosorbent assay in sera collected from preeclamptic pregnancies and gestation-age-matched controls. TGF-β signaling was assessed using an in vitro bioassay and a tube formation assay. RESULTS TGF-β type I, II, and III receptors were all identified in pregnant serum; all were substantially elevated in early-onset but not late-onset PE. Endoglin was increased in both subtypes. At the greatest concentrations detected in PE, none of these soluble TGF-β receptors alone, including endoglin, inhibited TGF-β signaling. However, when all four soluble receptors were present, signaling of both TGF-β1 and TGF-β2 was substantially reduced. Removal of any one of these soluble receptors alleviated TGF-β1 inhibition; however, removal of soluble TGFβRIII was necessary to relieve TGF-β2 inhibition. CONCLUSIONS Multiple soluble TGF-β receptors are present in pregnant circulation and elevated in early-onset PE; they synergistically inhibit TGF-β signaling, which might be more likely to occur in early-onset than late-onset PE. Reducing soluble TGFβRIII, rather than endoglin, would be more effective in alleviating the inhibition of both TGF-β1 and TGF-β2 signaling in PE.
Collapse
Affiliation(s)
- Yao Wang
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Qi Chen
- The Hospital of Obstetrics & Gynaecology, Fudan University, Shanghai 200090, China
- Department of Obstetrics & Gynaecology, The University of Auckland, Auckland 1142, New Zealand
| | - Min Zhao
- Wuxi Maternity and Children’s Health Hospital, Nanjing Medical University, Jiangsu 214002, China
| | - Kelly Walton
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
| | - Craig Harrison
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
| | - Guiying Nie
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
26
|
Filliat G, Mirsaidi A, Tiaden AN, Kuhn GA, Weber FE, Oka C, Richards PJ. Role of HTRA1 in bone formation and regeneration: In vitro and in vivo evaluation. PLoS One 2017; 12:e0181600. [PMID: 28732055 PMCID: PMC5521800 DOI: 10.1371/journal.pone.0181600] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/03/2017] [Indexed: 12/26/2022] Open
Abstract
The role of mammalian high temperature requirement protease A1 (HTRA1) in somatic stem cell differentiation and mineralized matrix formation remains controversial, having been demonstrated to impart either anti- or pro-osteogenic effects, depending on the in vitro cell model used. The aim of this study was therefore to further evaluate the role of HTRA1 in regulating the differentiation potential and lineage commitment of murine mesenchymal stem cells in vitro, and to assess its influence on bone structure and regeneration in vivo. Our results demonstrated that short hairpin RNA-mediated ablation of Htra1 in the murine mesenchymal cell line C3H10T1/2 increased the expression of several osteogenic gene markers, and significantly enhanced matrix mineralization in response to BMP-2 stimulation. These effects were concomitant with decreases in the expression of chondrogenic gene markers, and increases in adipogenic gene expression and lipid accrual. Despite the profound effects of loss-of-function of HTRA1 on this in vitro osteochondral model, these were not reproduced in vivo, where bone microarchitecture and regeneration in 16-week-old Htra1-knockout mice remained unaltered as compared to wild-type controls. By comparison, analysis of femurs from 52-week-old mice revealed that bone structure was better preserved in Htra1-knockout mice than age-matched wild-type controls. These findings therefore provide additional insights into the role played by HTRA1 in regulating mesenchymal stem cell differentiation, and offer opportunities for improving our understanding of how this multifunctional protease may act to influence bone quality.
Collapse
Affiliation(s)
- Gladys Filliat
- Bone and Stem Cell Research Group, CABMM, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Ali Mirsaidi
- Bone and Stem Cell Research Group, CABMM, University of Zurich, Zurich, Switzerland
| | - André N. Tiaden
- Bone and Stem Cell Research Group, CABMM, University of Zurich, Zurich, Switzerland
| | - Gisela A. Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Franz E. Weber
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- Oral Biotechnology & Bioengineering, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| | - Chio Oka
- Division of Gene Function in Animals, Nara Institute of Science and Technology, Nara, Japan
| | - Peter J. Richards
- Bone and Stem Cell Research Group, CABMM, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
27
|
Maternal HtrA3 optimizes placental development to influence offspring birth weight and subsequent white fat gain in adulthood. Sci Rep 2017; 7:4627. [PMID: 28676687 PMCID: PMC5496872 DOI: 10.1038/s41598-017-04867-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/22/2017] [Indexed: 12/25/2022] Open
Abstract
High temperature requirement factor A3 (HtrA3), a member of the HtrA protease family, is highly expressed in the developing placenta, including the maternal decidual cells in both mice and humans. In this study we deleted the HtrA3 gene in the mouse and crossed females carrying zero, one, or two HtrA3-expressing alleles with HtrA3+/− males to investigate the role of maternal vs fetal HtrA3 in placentation. Although HtrA3−/− mice were phenotypically normal and fertile, HtrA3 deletion in the mother resulted in intra-uterine growth restriction (IUGR). Disorganization of labyrinthine fetal capillaries was the major placental defect when HtrA3 was absent. The IUGR caused by maternal HtrA3 deletion, albeit being mild, significantly altered offspring growth trajectory long after birth. By 8 months of age, mice born to HtrA3-deficient mothers, independent of their own genotype, were significantly heavier and contained a larger mass of white fat. We further demonstrated that in women serum levels of HtrA3 during early pregnancy were significantly lower in IUGR pregnancies, establishing an association between lower HtrA3 levels and placental insufficiency in the human. This study thus revealed the importance of maternal HtrA3 in optimizing placental development and its long-term impact on the offspring well beyond in utero growth.
Collapse
|
28
|
Zurawa-Janicka D, Wenta T, Jarzab M, Skorko-Glonek J, Glaza P, Gieldon A, Ciarkowski J, Lipinska B. Structural insights into the activation mechanisms of human HtrA serine proteases. Arch Biochem Biophys 2017; 621:6-23. [PMID: 28396256 DOI: 10.1016/j.abb.2017.04.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022]
Abstract
Human HtrA1-4 proteins belong to the HtrA family of evolutionarily conserved serine proteases and function as important modulators of many physiological processes, including maintenance of mitochondrial homeostasis, cell signaling and apoptosis. Disturbances in their action are linked to severe diseases, including oncogenesis and neurodegeneration. The HtrA1-4 proteins share structural and functional features of other members of the HtrA protein family, however there are several significant differences in structural architecture and mechanisms of action which makes each of them unique. Our goal is to present recent studies regarding human HtrAs. We focus on their physiological functions, structure and regulation, and describe current models of activation mechanisms. Knowledge of molecular basis of the human HtrAs' action is a subject of great interest; it is crucial for understanding their relevance in cellular physiology and pathogenesis as well as for using them as targets in future therapies of diseases such as neurodegenerative disorders and cancer.
Collapse
Affiliation(s)
- Dorota Zurawa-Janicka
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland.
| | - Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Miroslaw Jarzab
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Joanna Skorko-Glonek
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Przemyslaw Glaza
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Artur Gieldon
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Jerzy Ciarkowski
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| |
Collapse
|
29
|
Wang Y, Li Y, Hyett J, da Silva Costa F, Nie G. HtrA3 Isoform-Specific ELISAs for Early Detection of Preeclampsia. SLAS DISCOVERY 2016; 23:1092-1099. [PMID: 27932697 DOI: 10.1177/1087057116682425] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Preeclampsia is a serious disorder of human pregnancy occurring after 20 weeks of gestation. It can be divided into subtypes of early onset (<34 weeks of gestation) and late onset (>34 weeks). Presymptomatic detection to identify those at high risk is important for managing this disease. HtrA3, a serine protease with high expression in the developing placenta, exists in long (HtrA3-L) and short (HtrA3-S) isoforms. They are identical, except HtrA3-S lacks the C-terminal PDZ domain. We have previously shown by Western blot analysis that serum HtrA3 levels at the end of the first trimester are significantly higher in women who later develop preeclampsia than in controls. In this study, using highly specific HtrA3 monoclonal antibodies, we established and fully validated two enzyme-linked immunosorbent assays to detect both HtrA3 isoforms together (HtrA3-T) and HtrA3-L alone in the human serum. We then determined serum HtrA3 at 11 to 13 weeks of gestation in a cohort of singleton pregnancies that proceeded without complications or developed preeclampsia in the third trimester. Compared with controls, those who developed late-onset preeclampsia had significantly higher levels of HtrA3-L, whereas those who developed early-onset preeclampsia had significantly lower ratios of HtrA3-L/HtrA3-T. These data support a potential utility of these HtrA3 ELISAs for early detection of preeclampsia.
Collapse
Affiliation(s)
- Yao Wang
- 1 Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia.,2 Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Ying Li
- 1 Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia.,2 Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Jonathan Hyett
- 3 Central Clinical School, University of Sydney, Sydney, NSW, Australia.,4 RPA Women and Babies, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Fabricio da Silva Costa
- 5 Monash Ultrasound for Women, Melbourne, VIC, Australia.,6 Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Guiying Nie
- 1 Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia.,2 Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia.,7 Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
30
|
Wang Y, Nie G. High levels of HtrA4 observed in preeclamptic circulation drastically alter endothelial gene expression and induce inflammation in human umbilical vein endothelial cells. Placenta 2016; 47:46-55. [PMID: 27780539 PMCID: PMC5090051 DOI: 10.1016/j.placenta.2016.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/31/2016] [Accepted: 09/07/2016] [Indexed: 01/14/2023]
Abstract
Introduction Preeclampsia (PE) is a life-threatening pregnancy disorder characterized by wide-spread endothelial dysfunction. Placental factors circulating in the maternal blood are believed to cause endothelial dysfunction. Our previous study identified HtrA4 as a placenta-specific serine protease that is released into the maternal circulation and significantly increased in early-onset PE. In this study, we examined the impact of HtrA4 on expression of endothelial genes related to vessel biology, using human umbilical vein endothelial cells (HUVECs) as a model. Methods HUVECs were treated with 0 or 3 μg/ml HtrA4 (highest concentration seen in PE circulation) for 24 h and analysed by an endothelial cell biology PCR array containing 84 genes. HtrA4-induced changes were then validated by real-time RT-PCR and ELISA for time and dose dependency. Results High levels of HtrA4 significantly altered the expression of a range of genes related to inflammation, vaso-activity, angiogenesis, cell adhesion, platelet activation and coagulation. In particular, pro-inflammatory genes IL6, PTGS2 (COX2) and IL1B were significantly increased by HtrA4. IL6 protein in HUVEC media was also drastically increased. THBD, an anticoagulant factor reported to be increased in PE, was significantly up-regulated by HtrA4. In contrast, THBS1, which is involved in many regulatory processes of endothelial cell biology, was severely down-regulated by HtrA4. Discussion HtrA4 significantly increased the inflammatory responses of HUVECs, and altered their expression of a number of genes important for vessel biology. These data suggest that placenta-derived HtrA4 that is increased in PE circulation is a potential causal factor of endothelial dysfunction. HtrA4 altered expression of endothelial genes involved in vessel biology. HtrA4 significantly induced inflammation in endothelial cells. Placental-derived HtrA4 is a potential causal factor of endothelial dysfunction.
Collapse
Affiliation(s)
- Yao Wang
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, 3800, Australia
| | - Guiying Nie
- Implantation and Placental Development Laboratory, Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
31
|
Serum levels of GDF15 are reduced in preeclampsia and the reduction is more profound in late-onset than early-onset cases. Cytokine 2016; 83:226-230. [DOI: 10.1016/j.cyto.2016.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/07/2016] [Accepted: 05/02/2016] [Indexed: 01/08/2023]
|