1
|
Zheng L, Tang R, Fang J, Hu H, Ahmad F, Tang Q, Liu J, Zhong M, Li J. Circular RNA hsa_circ_0081343 modulates trophoblast autophagy through Rbm8a nuclear translocation. Placenta 2024; 158:89-101. [PMID: 39413593 DOI: 10.1016/j.placenta.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/17/2024] [Accepted: 09/29/2024] [Indexed: 10/18/2024]
Abstract
INTRODUCTION Fetal growth restriction (FGR) is a kind of obstetric complication that seriously endangers fetal life. Recent studies reported significant reduction of hsa_circ_0081343 in human placenta developed in FGR and is involved in cell migration, invasion, and apoptosis of trophoblast by acting as microRNA sponges. Autophagy is required for invasion of trophoblast cells and for vascular remodeling during placentation. In this study, we aimed to explore the mechanistic link between hsa_circ_0081343 and autophagy. METHODS We investigated the interactions between hsa_circ_0081343 and RNA-binding proteins were studied by RNA pull-down assay, mass spectrometry and RNA immunoprecipitation assay. The mechanism of nuclear translocation of Rbm8a were assessed by reverse transcription-quantitative PCR, Western blot, immunofluorescence and Co-Immunoprecipitation. Western blot, immunofluorescence and transmission electron microscopy were performed to elucidate the mechanism underlying hsa_circ_0081343 and/or Rbm8a mediated regulation of autophagy. RESULTS hsa_circ_0081343 served as an RNA-binding protein (RBP) sponge. RNA binding motif protein 8A (Rbm8a) was directly bound to hsa_circ_0081343 in the cytoplasm, while knockdown of hsa_circ_0081343 facilitated Rbm8a localization in the nucleus. We also identified Rbm8a as a potential import cargo for Importin13 (Ipo13), which transported Rbm8a across the nuclear membrane into the nucleus. Ipo13 recognized Rbm8a via a functional nuclear localization signal (NLS). Furthermore, the mechanistic study revealed that hsa_circ_0081343-mediated nuclear translocation of Rbm8a activated trophoblast autophagy. DISCUSSION Our results suggest that hsa_circ_0081343 could bind to RBP and the interaction between hsa_circ_0081343 and Rbm8a participate in regulating autophagy. These findings offer novel molecular targets and insights for a potential therapeutic strategy against FGR.
Collapse
Affiliation(s)
- Linmei Zheng
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China; Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Rong Tang
- Department of Hepatological Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Junbo Fang
- Department of Pathology, Southern Medical University, Guangzhou, 510515, China
| | - Haoyue Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Fiaz Ahmad
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University (NPU), Xi'an, 710072, Shaanxi, China
| | - Qiong Tang
- Department of Pathology, Southern Medical University, Guangzhou, 510515, China
| | - Jinfu Liu
- Department of Pathology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Jing Li
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Shen Q, Yu L, Zhang H. Internal Quality Network Stress and Inflammation Reaction Indicators in the Expression and Significance of the Serum of Limited Pregnant Women's Serum. Br J Hosp Med (Lond) 2024; 85:1-13. [PMID: 39618207 DOI: 10.12968/hmed.2024.0399] [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] [Indexed: 02/05/2025]
Abstract
Aims/Background Fetal growth restriction (FGR) is a common clinical disorder in pregnant women. Inflammation and endoplasmic reticulum stress play an important role in the occurrence and development of FGR. The purpose of this study was to explore the expression and significance of endoplasmic reticulum stress and inflammatory response indicators in the serum of pregnant women with FGR. Methods The data of pregnant women admitted to Changzhou Maternal and Child Health Care Hospital from January 2020 to June 2023 were collected and analyzed by propensity score matching (PSM). Pregnant women with FGR were included in the observation group (n = 65), whereas healthy pregnant women admitted to the hospital during the same period were included in the control group (n = 65). Enzyme-linked immunosorbent assay (ELISA) was used to detect the serum levels of inflammatory markers such as tumor necrosis factor alpha (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) in all the pregnant women recruited in this study. The serum levels of glucose regulatory protein 78 (GRP78) and C/EBP homologous protein (CHOP), which are characteristic indicators of endoplasmic reticulum stress, in the two groups of pregnant women, were also detected and analyzed. A range of parameters concerning fetal growth and development indicators, such as fetal head circumference, abdominal circumference and fetal development index, of the two groups of study subjects were recorded and compared. Analyze the correlation between inflammatory response indicators and endoplasmic reticulum stress factors in pregnant women with FGR and fetal growth and development indicators. Results The serum levels of TNF-α, IL-1β and IL-6 in the observation group were significantly higher than those in the control group (p < 0.001). The serum levels of GRP78 and CHOP in the observation group were significantly higher than those in the control group (p < 0.001). The levels of GRP78 and CHOP in pregnant women with FGR were negatively correlated with fetal head circumference, abdominal circumference, and fetal growth and development index (p < 0.05). Conclusion The serum levels of inflammatory markers such as TNF-α, IL-1β and IL-6 in pregnant women with FGR were abnormally elevated, indicating severe inflammatory response. In addition, endoplasmic reticulum stress was observed in pregnant women with FGR, marked by significantly elevated levels of GRP78 and CHOP. The levels of TNF-α, IL-1β, IL-6, GRP78 and CHOP were negatively correlated with fetal head circumference, abdominal circumference and fetal growth and development index, implying their impacts on the occurrence and development of FGR.
Collapse
Affiliation(s)
- Qi Shen
- Department of Obstetrics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Litao Yu
- Department of Obstetrics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hui Zhang
- Department of Clinical Laboratory, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
3
|
Yang T, Hu J, Zhang L, Liu L, Pan X, Zhou Y, Wu Y, Shi X, Obiegbusi CN, Dong X. CircCUL1 inhibits trophoblast cell migration and invasion and promotes cell autophagy by sponging hsa-miR-30e-3p in fetal growth restriction via the ANXA1/PI3K/AKT axis. J Biochem Mol Toxicol 2024; 38:e23759. [PMID: 39003567 DOI: 10.1002/jbt.23759] [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: 03/01/2023] [Revised: 03/26/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Fetal growth restriction (FGR) severely affects the health outcome of newborns and represents a major cause of perinatal morbidity. The precise involvement of circCULT1 in the progression of FGR remains unclear. We performed next-generation sequencing and RT-qPCR to identify differentially expressed circRNAs in placental tissues affected by FGR by comparing them with unaffected counterparts. Edu, flow cytometry, and transwell assay were conducted to detect HTR8/SVneo cell's function in regard to cell proliferation, migration, and invasion. The interaction between circCUL1 and hsa-miR-30e-3p was assessed through dual-luciferase reporter assays, validation of the interaction between circCUL1 and ANXA1 was performed using RNA pulldown and immunoprecipitation assays. Western blot analysis was performed to evaluate protein levels of autophagy markers and components of the PI3K/AKT signaling pathway. A knockout (KO) mouse model was established for homologous mmu-circ-0001469 to assess fetal mouse growth and development indicators. Our findings revealed an upregulation of circCUL1 expression in placental tissues from patients with FGR. We found that suppression of circCUL1 increased the trophoblast cell proliferation, migration, and invasion, circCUL1 could interact with hsa-miR-30e-3p. Further, circCUL1 stimulated autophagy, modulating trophoblast cell autophagy via the ANXA1/PI3K/AKT pathway, and a notable disparity was observed, with KO mice displaying accelerated embryo development and exhibiting heavier placentas in comparison to wild-type C57BL/6 mice. By modulating the ANXA1/PI3K/AKT signaling pathway through the interaction with hsa-miR-30e-3p, circCUL1 promotes autophagy while concurrently suppressing trophoblast cell proliferation, migration, and invasion. These findings offer novel insights into potential diagnostic markers and therapeutic targets for FGR research.
Collapse
Affiliation(s)
- Tong Yang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jianguo Hu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lei Zhang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Li Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xin Pan
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yanqiu Zhou
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yi Wu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xian Shi
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Chidera N Obiegbusi
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xiaojing Dong
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Zhang YF, Zhang S, Ling Q, Chang W, Tan LL, Zhang J, Xiong YW, Zhu HL, Bian P, Wang H. Activation of lipophagy ameliorates cadmium-induced neural tube defects via reducing low density lipoprotein cholesterol levels in mouse placentas. Cell Biol Toxicol 2024; 40:35. [PMID: 38771546 PMCID: PMC11108957 DOI: 10.1007/s10565-024-09885-2] [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: 02/22/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Neural tube defects (NTDs) represent a prevalent and severe category of congenital anomalies in humans. Cadmium (Cd) is an environmental teratogen known to cause fetal NTDs. However, its underlying mechanisms remain elusive. This study aims to investigate the therapeutic potential of lipophagy in the treatment of NTDs, providing valuable insights for future strategies targeting lipophagy activation as a means to mitigate NTDs.We successfully modeled NTDs by Cd exposure during pregnancy. RNA sequencing was employed to investigate the transcriptomic alterations and functional enrichment of differentially expressed genes in NTD placental tissues. Subsequently, pharmacological/genetic (Atg5-/- placentas) experiments confirmed that inducing placental lipophagy can alleviate Cd induced-NTDs. We found that Cd exposure caused NTDs. Further analyzed transcriptomic data from the placentas with NTDs which revealed significant downregulation of low-density lipoprotein receptor associated protein 1(Lrp1) gene expression responsible for positive regulation of low-density lipoprotein cholesterol (LDL-C) transport. Correspondingly, there was an increase in maternal serum/placenta/amniotic fluid LDL-C content. Subsequently, we have discovered that Cd exposure activated placental lipophagy. Pharmacological/genetic (Atg5-/- placentas) experiments confirmed that inducing placental lipophagy can alleviate Cd induced-NTDs. Furthermore, our findings demonstrate that activation of placental lipophagy effectively counteracts the Cd-induced elevation in LDL-C levels. Lipophagy serves to mitigate Cd-induced NTDs by reducing LDL-C levels within mouse placentas.
Collapse
Affiliation(s)
- Yu-Feng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Shuang Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Qing Ling
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Wei Chang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Lu-Lu Tan
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Jin Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Yong-Wei Xiong
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Hua-Long Zhu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Po Bian
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China.
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China.
| |
Collapse
|
5
|
Dai Y, Sang XB, Bai WP. N-acetylcysteine and Hydroxychloroquine Ameliorate ADMA-Induced Fetal Growth Restriction in Mice via Regulating Oxidative Stress and Autophagy. Reprod Sci 2024; 31:779-790. [PMID: 37845590 DOI: 10.1007/s43032-023-01380-z] [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: 09/12/2023] [Accepted: 10/03/2023] [Indexed: 10/18/2023]
Abstract
Fetal growth restriction (FGR) seriously threatens perinatal health. The main cause of FGR is placental malperfusion, but the specific mechanism is still unclear, and there is no effective treatment for FGR. We constructed a FGR mouse model by adding exogenous asymmetric dimethylarginine (ADMA) through in vivo experiments and found that ADMA could cause placental dysplasia and induce the occurrence of FGR. Compared with the control group, reactive oxygen species (ROS) production in the placenta was increased in mice with FGR, and the expression of autophagy-related proteins p-AKT/AKT, p-mTOR/mTOR, and P62 was significantly decreased, while the expression of Beclin-1 and LC3-II was significantly increased in the FGR group. Furthermore, ADMA had a favorable effect in promoting the formation of autophagosomes. Hydroxychloroquine (HCQ) and N-acetylcysteine (NAC) improved ADMA-induced disorders of placental development and alleviated ADMA-induced FGR. This study found that ADMA could cause excessive autophagy of trophoblasts by increasing the level of oxidative stress, ultimately leading to the occurrence of FGR, and HCQ and NAC had therapeutic effects on ADMA-induced FGR.
Collapse
Affiliation(s)
- Yan Dai
- The Department of Gynecology and Obstetrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiu-Bo Sang
- The Department of Gynecology and Obstetrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wen-Pei Bai
- The Department of Gynecology and Obstetrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Zhou P, Wang J, Wang J, Liu X. When autophagy meets placenta development and pregnancy complications. Front Cell Dev Biol 2024; 12:1327167. [PMID: 38371923 PMCID: PMC10869551 DOI: 10.3389/fcell.2024.1327167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/22/2024] [Indexed: 02/20/2024] Open
Abstract
Autophagy is a common biological phenomenon in eukaryotes that has evolved and reshaped to maintain cellular homeostasis. Under the pressure of starvation, hypoxia, and immune damage, autophagy provides energy and nutrients to cells, which benefits cell survival. In mammals, autophagy is an early embryonic nutrient supply system involved in early embryonic development, implantation, and pregnancy maintenance. Recent studies have found that autophagy imbalance in placental tissue plays a key role in the occurrence and development of pregnancy complications, such as gestational hypertension, gestational obesity, premature birth, miscarriage, and intrauterine growth restriction. This mini-review summarizes the molecular mechanism of autophagy regulation, the autophagy pathways, and related factors involved in placental tissue and comprehensively describes the role of autophagy in pregnancy complications.
Collapse
Affiliation(s)
- Pei Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Junqi Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jun Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Obstetrics and Gynecology, Benxi Central Hospital of China Medical University, Benxi, Liaoning, China
| | - Xiaomei Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
7
|
Carvajal L, Gutiérrez J, Morselli E, Leiva A. Autophagy Process in Trophoblast Cells Invasion and Differentiation: Similitude and Differences With Cancer Cells. Front Oncol 2021; 11:637594. [PMID: 33937039 PMCID: PMC8082112 DOI: 10.3389/fonc.2021.637594] [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: 12/03/2020] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Early human placental development begins with blastocyst implantation, then the trophoblast differentiates and originates the cells required for a proper fetal nutrition and placental implantation. Among them, extravillous trophoblast corresponds to a non-proliferating trophoblast highly invasive that allows the vascular remodeling which is essential for appropriate placental perfusion and to maintain the adequate fetal growth. This process involves different placental cell types as well as molecules that allow cell growth, cellular adhesion, tissular remodeling, and immune tolerance. Remarkably, some of the cellular processes required for proper placentation are common between placental and cancer cells to finally support tumor growth. Indeed, as in placentation trophoblasts invade and migrate, cancer cells invade and migrate to promote tumor metastasis. However, while these processes respond to a controlled program in trophoblasts, in cancer cells this regulation is lost. Interestingly, it has been shown that autophagy, a process responsible for the degradation of damaged proteins and organelles to maintain cellular homeostasis, is required for invasion of trophoblast cells and for vascular remodeling during placentation. In cancer cells, autophagy has a dual role, as it has been shown both as tumor promoter and inhibitor, depending on the stage and tumor considered. In this review, we summarized the similarities and differences between trophoblast cell invasion and cancer cell metastasis specifically evaluating the role of autophagy in both processes.
Collapse
Affiliation(s)
- Lorena Carvajal
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jaime Gutiérrez
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago, Chile
| | - Eugenia Morselli
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,Autophagy Research Center, Santiago, Chile
| | - Andrea Leiva
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago, Chile
| |
Collapse
|