1
|
Li M, Wang L, Luo Y, Wang J, Liu X, Li S, Hao Z. RNA-seq analysis of the biological process and regulatory signal of TGF-β1-mediated changes in ovarian granulosa cells in small-tail Han sheep. Theriogenology 2025; 234:9-18. [PMID: 39631254 DOI: 10.1016/j.theriogenology.2024.12.001] [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: 08/20/2024] [Revised: 11/25/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Transforming growth factor beta-1 (TGF-β1) regulates the proliferation of ovarian granulosa cells and participates in follicular development in small-tail Han sheep via the SMAD pathway. However, which additional biological processes and regulatory mechanisms are involved in TGF-β1-mediated regulation of granulosa cell changes remains unknown. In this study, TGF-β1-treated (10 ng/mL) ovarian granulosa cells of small-tail Han sheep were used as the model, RNA-Seq was employed to screen differentially expressed genes (DEGs), and rescue experiments were used to verify selected key pathways. In total, 1179 upregulated and 873 downregulated DEGs were screened using RNA-Seq. Gene Ontology (GO) enrichment analysis showed that the DEGs were mainly involved in the biological processes of cell adhesion, cell migration, cell cycle, cell proliferation and apoptosis, and endocrine regulation. Meanwhile, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that the DEGs were primarily associated with pathways relating to ECM-receptor interaction, PI3K-AKT, focal adhesion, MAPK, TNF, and FOXO signaling, among others. The addition of doramapimod confirmed that the p38 pathway participates in the TGF-β1-induced proliferation and apoptosis of ovarian granulosa cells as well as the regulation of steroid hormone secretion. These results revealed a novel TGF-β1/p38 pathway-mediated mechanism that induces both the proliferation and apoptosis of ovarian granulosa cells. Our findings provide a basis for better understanding the genetic mechanism of TGF-β1 action in follicle development.
Collapse
Affiliation(s)
- Mingna Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Engineering Lab of Genetic Improvement in Ruminants, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Longbin Wang
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Engineering Lab of Genetic Improvement in Ruminants, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Yuzhu Luo
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Engineering Lab of Genetic Improvement in Ruminants, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jiqing Wang
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Engineering Lab of Genetic Improvement in Ruminants, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Xiu Liu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Engineering Lab of Genetic Improvement in Ruminants, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Shaobin Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Engineering Lab of Genetic Improvement in Ruminants, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhiyun Hao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Engineering Lab of Genetic Improvement in Ruminants, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| |
Collapse
|
2
|
Lv R, Yao Y, Dong J, Chen Q. COL1A1, mediated by m6A methylation of METTL3, facilitates oral squamous cell carcinoma cell growth and metastasis. Odontology 2025; 113:191-200. [PMID: 38900231 DOI: 10.1007/s10266-024-00962-w] [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: 12/07/2023] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Collagen type I alpha1 (COL1A1) has been found to be abnormal expressed in oral squamous cell carcinoma (OSCC) tissues, but its role and mechanism in OSCC need to be further elucidated. The expression levels of COL1A1 and methyltransferase-like 3 (METTL3) were measured by quantitative real-time PCR and western blot. Cell growth and metastasis were determined by CCK8, colony formation, EdU, flow cytometry and transwell assays. MeRIP, Co-IP and dual-luciferase reporter assays were performed to explore the interplay of COL1A1 and METTL3. COL1A1 mRNA stability was confirmed by Actinomycin D assay. Mice xenograft models were constructed to perform in vivo experiments. COL1A1 and METTL3 were upregulated in OSCC. COL1A1 knockdown suppressed OSCC cell growth and metastasis, while its overexpression had an opposite effect. The stability of COL1A1 mRNA was regulated by the m6A methylation of METTL3. METTL3 overexpression promoted OSCC cell growth and metastasis, and its knockdown-mediated OSCC cell function inhibition could be abolished by COL1A1 overexpression. Besides, silencing of METTL3 reduced OSCC tumor growth by reducing COL1A1 expression. METTL3-stabilized COL1A1 promoted OSCC progression, providing an exact molecular target for the treatment of OSCC.
Collapse
Affiliation(s)
- Ruya Lv
- Department of Stomatology, Jingzhou Central Hospital, No. 6 Jingzhong Road, Jingzhou District, Jingzhou, 434000, Hubei, China.
- Department of Stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, Hubei, China.
| | - Yao Yao
- Department of Stomatology, Jingzhou Central Hospital, No. 6 Jingzhong Road, Jingzhou District, Jingzhou, 434000, Hubei, China
- Department of Stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, Hubei, China
| | - Jingjing Dong
- Department of Stomatology, Jingzhou Central Hospital, No. 6 Jingzhong Road, Jingzhou District, Jingzhou, 434000, Hubei, China
- Department of Stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, Hubei, China
| | - Qian Chen
- Department of Stomatology, Jingzhou Central Hospital, No. 6 Jingzhong Road, Jingzhou District, Jingzhou, 434000, Hubei, China
- Department of Stomatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, Hubei, China
| |
Collapse
|
3
|
Ruggeri E, Klohonatz K, Durrant B, Sirard MA. Identification and Preliminary Analysis of Granulosa Cell Biomarkers to Predict Oocyte In Vitro Maturation Outcome in the Southern White Rhinoceros ( Ceratotherium simum simum). Animals (Basel) 2024; 14:3538. [PMID: 39682503 DOI: 10.3390/ani14233538] [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: 11/11/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
In recent years, biomarkers in granulosa cells (GC) have been determined and associated in several species with oocyte maturation, in vitro fertilization success, and embryo development outcomes. The identification of biomarkers of oocyte competence can aid in improving assisted reproductive technologies (ARTs) in the southern white rhino (SWR). This study aimed to identify biomarkers present in SWR GC associated with oocytes that either did or did not mature in vitro. We evaluated follicle development (FD), meiotic competence (MC), cell death and atresia (CDA), and embryonic genome activation (EGA). Our objective was to design biomarkers to predict oocyte in vitro maturation results in the SWR. RNA was isolated from GC obtained during ovum pick up (OPU) for qPCR analysis. Overall, 22 genes were assessed, and nine were differentially expressed between GC from oocytes that did or did not mature in vitro (FD-GDF9 and mTOR; MC-GGPS1, JMY, and NPR2; CDA-COL4A1, MACIR, and TMPO; EGA-NFYA). From these data, we determined that GC can be used as a predictor for oocyte in vitro maturation outcome in the SWR. Our results provide crucial information needed to improve in vitro maturation and ARTs in this species.
Collapse
Affiliation(s)
- Elena Ruggeri
- Reproductive Sciences, Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, CA 92027, USA
| | - Kristin Klohonatz
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Barbara Durrant
- Reproductive Sciences, Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, CA 92027, USA
| | - Marc-André Sirard
- Département des Sciences Animales, Université Laval, Quebec City, QC G1V 0A6, Canada
| |
Collapse
|
4
|
Guo Y, Xue L, Tang W, Xiong J, Chen D, Dai Y, Wu C, Wei S, Dai J, Wu M, Wang S. Ovarian microenvironment: challenges and opportunities in protecting against chemotherapy-associated ovarian damage. Hum Reprod Update 2024; 30:614-647. [PMID: 38942605 PMCID: PMC11369228 DOI: 10.1093/humupd/dmae020] [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: 10/30/2023] [Revised: 04/27/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Chemotherapy-associated ovarian damage (CAOD) is one of the most feared short- and long-term side effects of anticancer treatment in premenopausal women. Accumulating detailed data show that different chemotherapy regimens can lead to disturbance of ovarian hormone levels, reduced or lost fertility, and an increased risk of early menopause. Previous studies have often focused on the direct effects of chemotherapeutic drugs on ovarian follicles, such as direct DNA damage-mediated apoptotic death and primordial follicle burnout. Emerging evidence has revealed an imbalance in the ovarian microenvironment during chemotherapy. The ovarian microenvironment provides nutritional support and transportation of signals that stimulate the growth and development of follicles, ovulation, and corpus luteum formation. The close interaction between the ovarian microenvironment and follicles can determine ovarian function. Therefore, designing novel and precise strategies to manipulate the ovarian microenvironment may be a new strategy to protect ovarian function during chemotherapy. OBJECTIVE AND RATIONALE This review details the changes that occur in the ovarian microenvironment during chemotherapy and emphasizes the importance of developing new therapeutics that protect ovarian function by targeting the ovarian microenvironment during chemotherapy. SEARCH METHODS A comprehensive review of the literature was performed by searching PubMed up to April 2024. Search terms included 'ovarian microenvironment' (ovarian extracellular matrix, ovarian stromal cells, ovarian interstitial, ovarian blood vessels, ovarian lymphatic vessels, ovarian macrophages, ovarian lymphocytes, ovarian immune cytokines, ovarian oxidative stress, ovarian reactive oxygen species, ovarian senescence cells, ovarian senescence-associated secretory phenotypes, ovarian oogonial stem cells, ovarian stem cells), terms related to ovarian function (reproductive health, fertility, infertility, fecundity, ovarian reserve, ovarian function, menopause, decreased ovarian reserve, premature ovarian insufficiency/failure), and terms related to chemotherapy (cyclophosphamide, lfosfamide, chlormethine, chlorambucil, busulfan, melphalan, procarbazine, cisplatin, doxorubicin, carboplatin, taxane, paclitaxel, docetaxel, 5-fluorouraci, vincristine, methotrexate, dactinomycin, bleomycin, mercaptopurine). OUTCOMES The ovarian microenvironment shows great changes during chemotherapy, inducing extracellular matrix deposition and stromal fibrosis, angiogenesis disorders, immune microenvironment disturbance, oxidative stress imbalances, ovarian stem cell exhaustion, and cell senescence, thereby lowering the quantity and quality of ovarian follicles. Several methods targeting the ovarian microenvironment have been adopted to prevent and treat CAOD, such as stem cell therapy and the use of free radical scavengers, senolytherapies, immunomodulators, and proangiogenic factors. WIDER IMPLICATIONS Ovarian function is determined by its 'seeds' (follicles) and 'soil' (ovarian microenvironment). The ovarian microenvironment has been reported to play a vital role in CAOD and targeting the ovarian microenvironment may present potential therapeutic approaches for CAOD. However, the relation between the ovarian microenvironment, its regulatory networks, and CAOD needs to be further studied. A better understanding of these issues could be helpful in explaining the pathogenesis of CAOD and creating innovative strategies for counteracting the effects exerted on ovarian function. Our aim is that this narrative review of CAOD will stimulate more research in this important field. REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Yican Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Yun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Chuqing Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Simin Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| |
Collapse
|
5
|
Klohonatz K, Durrant B, Sirard MA, Ruggeri E. Granulosa cells provide transcriptomic information on ovarian follicle dynamics in southern white rhinoceros. Sci Rep 2024; 14:19321. [PMID: 39164442 PMCID: PMC11336098 DOI: 10.1038/s41598-024-70235-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
Much remains unknown about the reproductive physiology of southern white rhinoceros (SWR) and the effect of ovarian stimulation prior to ovum pickup (OPU) have not been fully elucidated. Granulosa cells (GC) provide valuable insight into follicle growth and oocyte maturation status. The goals of this study were to evaluate transcriptomic changes in GC from three stages of follicle development and to identify biomarkers possibly associated with follicular growth and maturation as a result of ovarian stimulation. GC collected from SWRs following OPU were assigned stages based upon follicle size. Total RNA was isolated, and cDNA libraries were prepared and sequenced on a NovaSeq 6000. All bioinformatics analyses were performed utilizing the Galaxy web platform. Reads were aligned to CerSimCot1.0, and the manual curation was performed with EquCab3.0. Overall, 39,455 transcripts (21,612 genes) were identified across follicle stages, and manual curation yielded a 61% increase in gene identification from the original annotation. Granulosa cells from preovulatory follicles expressed the highest number of unique transcripts. The following seven biomarkers were determined based upon cluster analysis and patterns of expression: COL1A1, JMY, FBXW11, NRG1, TMPO, MACIR and COL4A1. These data can be used to potentially evaluate the effects of different ovarian stimulation protocols on follicle dynamics, improve OPU results, and support conservation efforts in this species.
Collapse
Affiliation(s)
- Kristin Klohonatz
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Barbara Durrant
- Reproductive Sciences, Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, CA, USA
| | - Marc-André Sirard
- Département des Sciences Animales, Université Laval, Québec City, Québec, Canada
| | - Elena Ruggeri
- Reproductive Sciences, Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, CA, USA.
| |
Collapse
|
6
|
Sugioka K, Nishida T, Murakami J, Itahashi M, Yunoki M, Kusaka S. Substance P promotes transforming growth factor-β-induced collagen synthesis in human corneal fibroblasts. Am J Physiol Cell Physiol 2024; 326:C1482-C1493. [PMID: 38525537 DOI: 10.1152/ajpcell.00084.2024] [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: 02/04/2024] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
Corneal fibroblasts maintain homeostasis of the corneal stroma by mediating the synthesis and degradation of extracellular collagen, and these actions are promoted by transforming growth factor-β (TGF-β) and interleukin-1β (IL-1β), respectively. The cornea is densely innervated with sensory nerve fibers that are not only responsible for sensation but also required for physiological processes such as tear secretion and wound healing. Loss or dysfunction of corneal nerves thus impairs corneal epithelial wound healing and can lead to neurotrophic keratopathy. The sensory neurotransmitter substance P (SP) promotes corneal epithelial wound healing by enhancing the stimulatory effects of growth factors and fibronectin. We have now investigated the role of SP in collagen metabolism mediated by human corneal fibroblasts in culture. Although SP alone had no effect on collagen synthesis or degradation by these cells, it promoted the stimulatory effect of TGF-β on collagen type I synthesis without affecting that of IL-1β on the expression of matrix metalloproteinase-1. This effect of SP on TGF-β-induced collagen synthesis was accompanied by activation of p38 mitogen-activated protein kinase (MAPK) signaling and was attenuated by pharmacological inhibition of p38 or of the neurokinin-1 receptor. Our results thus implicate SP as a modulator of TGF-β-induced collagen type I synthesis by human corneal fibroblasts, and they suggest that loss of this function may contribute to the development of neurotrophic keratopathy.NEW & NOTEWORTHY This study investigates the role of substance P (SP) in collagen metabolism mediated by human corneal fibroblasts in culture. We found that, although SP alone had no effect on collagen synthesis or degradation by corneal fibroblasts, it promoted the stimulatory effect of transforming growth factor-β on collagen type I synthesis without affecting that of interleukin-1β on the expression of matrix metalloproteinase-1.
Collapse
Affiliation(s)
- Koji Sugioka
- Department of Ophthalmology, Kindai University Nara Hospital, Ikoma, Japan
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Teruo Nishida
- Department of Ophthalmology, Kindai University Nara Hospital, Ikoma, Japan
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Japan
- Division of Cornea and Ocular Surface, Ohshima Eye Hospital, Fukuoka, Japan
| | | | | | - Mai Yunoki
- Department of Ophthalmology, Kindai University Nara Hospital, Ikoma, Japan
| | - Shunji Kusaka
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osakasayama, Japan
| |
Collapse
|
7
|
Li M, Liang W, Luo Y, Wang J, Liu X, Li S, Hao Z. Transforming growth factor-β1 mediates the SMAD4/BMF pathway to regulate ovarian granulosa cell apoptosis in small tail Han sheep. Theriogenology 2024; 214:360-369. [PMID: 37979327 DOI: 10.1016/j.theriogenology.2023.11.009] [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: 08/07/2023] [Revised: 10/23/2023] [Accepted: 11/08/2023] [Indexed: 11/20/2023]
Abstract
Transforming growth factor (TGF)-β1 is an important multifunctional cytokine in the TGF-β signaling pathway, which is involved in the molecular regulation of multiple activities, including follicle development and ovulation in female reproductive physiology. However, the biological function of TGF-β1 in follicular development and in regulating the proliferation or apoptosis of granulosa cells in small tail Han sheep remain unclear. In this study, we analyzed the expression levels of TGF-β1 in the ovary at the follicular stage in small tail Han sheep. We further examined the effects of TGF-β1 on the viability, proliferation, and apoptosis of granulosa cells. Differential expression of TGF-β1 at the mRNA and protein levels was detected in the ovaries between the beginning of estrus and at preovulation. Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine (EdU) labeling, and flow cytometry assays showed that adding 5 and 10 ng/mL TGF-β1 could improve the viability and growth rate, reduce the apoptosis rate, and reduce the expression level of the pro-apoptotic factor Bcl-2-modifying factor (BMF) in granulosa cells. Treatment of 10 ng/mL TGF-β1 at all time points (except 72 h) significantly increased the positive rate of EdU labeling compared to that of the control group. RNA interference of SMAD4 reversed the decreased apoptosis rate caused by stimulation with 10 ng/mL TGF-β1, accompanied by a corresponding increase in the BMF expression level. Collectively, these results indicate that TGF-β1 plays a role in the ovarian follicular-phase activity of small tail Han sheep by inhibiting the apoptosis of sheep granulosa cells through the SMAD4/BMF pathway to promote proliferation and vitality. This study provides new insight into the molecular mechanism underlying TGF-β1 function regulation in granulosa cells, suggests a new target for the regulation of follicle development, and expands the new field of animal reproduction regulation technology.
Collapse
Affiliation(s)
- Mingna Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Weiwei Liang
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Yuzhu Luo
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jiqing Wang
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Xiu Liu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Shaobin Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhiyun Hao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| |
Collapse
|
8
|
Devos H, Zoidakis J, Roubelakis MG, Latosinska A, Vlahou A. Reviewing the Regulators of COL1A1. Int J Mol Sci 2023; 24:10004. [PMID: 37373151 DOI: 10.3390/ijms241210004] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/28/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
The collagen family contains 28 proteins, predominantly expressed in the extracellular matrix (ECM) and characterized by a triple-helix structure. Collagens undergo several maturation steps, including post-translational modifications (PTMs) and cross-linking. These proteins are associated with multiple diseases, the most pronounced of which are fibrosis and bone diseases. This review focuses on the most abundant ECM protein highly implicated in disease, type I collagen (collagen I), in particular on its predominant chain collagen type I alpha 1 (COLα1 (I)). An overview of the regulators of COLα1 (I) and COLα1 (I) interactors is presented. Manuscripts were retrieved searching PubMed, using specific keywords related to COLα1 (I). COL1A1 regulators at the epigenetic, transcriptional, post-transcriptional and post-translational levels include DNA Methyl Transferases (DNMTs), Tumour Growth Factor β (TGFβ), Terminal Nucleotidyltransferase 5A (TENT5A) and Bone Morphogenic Protein 1 (BMP1), respectively. COLα1 (I) interacts with a variety of cell receptors including integrinβ, Endo180 and Discoidin Domain Receptors (DDRs). Collectively, even though multiple factors have been identified in association to COLα1 (I) function, the implicated pathways frequently remain unclear, underscoring the need for a more spherical analysis considering all molecular levels simultaneously.
Collapse
Affiliation(s)
- Hanne Devos
- Centre of Systems Biology, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Jerome Zoidakis
- Centre of Systems Biology, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Maria G Roubelakis
- Laboratory of Biology, University of Athens School of Medicine, 11527 Athens, Greece
- Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| | | | - Antonia Vlahou
- Centre of Systems Biology, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
9
|
Xia L, Shen Y, Liu S, Du J. Iron overload triggering ECM-mediated Hippo/YAP pathway in follicle development: a hypothetical model endowed with therapeutic implications. Front Endocrinol (Lausanne) 2023; 14:1174817. [PMID: 37223010 PMCID: PMC10200985 DOI: 10.3389/fendo.2023.1174817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/12/2023] [Indexed: 05/25/2023] Open
Abstract
Disruption of iron homeostasis plays a negative role in follicle development. The dynamic changes in follicle growth are dependent on Hippo/YAP signaling and mechanical forces. However, little is known about the liaison between iron overload and the Hippo/YAP signalling pathway in term of folliculogenesis. Here, based on the available evidence, we established a hypothesized model linking excessive iron, extracellular matrix (ECM), transforming growth factor-β (TGF-β) and Hippo/Yes-associated protein (YAP) signal regarding follicle development. Hypothetically, the TGF-β signal and iron overload may play a synergistic role in ECM production via YAP. We speculate that the dynamic homeostasis of follicular iron interacts with YAP, increasing the risk of ovarian reserve loss and may enhance the sensitivity of follicles to accumulated iron. Hence, therapeutic interventions targeting iron metabolism disorders, and Hippo/YAP signal may alter the consequences of the impaired developmental process based on our hypothesis, which provides potential targets and inspiration for further drug discovery and development applied to clinical treatment.
Collapse
Affiliation(s)
- Lingjin Xia
- National Health Commission of the People's Republic of China (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Yupei Shen
- National Health Commission of the People's Republic of China (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Suying Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Du
- National Health Commission of the People's Republic of China (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Zhao J, Pan H, Liu Y, He Y, Shi H, Ge C. Interacting Networks of the Hypothalamic-Pituitary-Ovarian Axis Regulate Layer Hens Performance. Genes (Basel) 2023; 14:141. [PMID: 36672882 PMCID: PMC9859134 DOI: 10.3390/genes14010141] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Egg production is a vital biological and economic trait for poultry breeding. The 'hypothalamic-pituitary-ovarian (HPO) axis' determines the egg production, which affects the layer hens industry income. At the organism level, the HPO axis is influenced by the factors related to metabolic and nutritional status, environment, and genetics, whereas at the cellular and molecular levels, the HPO axis is influenced by the factors related to endocrine and metabolic regulation, cytokines, key genes, signaling pathways, post-transcriptional processing, and epigenetic modifications. MiRNAs and lncRNAs play a critical role in follicle selection and development, atresia, and ovulation in layer hens; in particular, miRNA is known to affect the development and atresia of follicles by regulating apoptosis and autophagy of granulosa cells. The current review elaborates on the regulation of the HPO axis and its role in the laying performance of hens at the organism, cellular, and molecular levels. In addition, this review provides an overview of the interactive network regulation mechanism of the HPO axis in layer hens, as well as comprehensive knowledge for successfully utilizing their genetic resources.
Collapse
Affiliation(s)
- Jinbo Zhao
- Faculty of Animal Science and Technology, Yunnan Agricultural University Kunming, Kunming 650201, China
- Branch of Animal Husbandry and Veterinary of Heilongjiang Academy of Agricultural Sciences, Qiqihar 161005, China
| | - Hongbin Pan
- Faculty of Animal Science and Technology, Yunnan Agricultural University Kunming, Kunming 650201, China
| | - Yong Liu
- Faculty of Animal Science and Technology, Yunnan Agricultural University Kunming, Kunming 650201, China
| | - Yang He
- Faculty of Animal Science and Technology, Yunnan Agricultural University Kunming, Kunming 650201, China
| | - Hongmei Shi
- Faculty of Animal Science and Technology, Yunnan Agricultural University Kunming, Kunming 650201, China
| | - Changrong Ge
- Faculty of Animal Science and Technology, Yunnan Agricultural University Kunming, Kunming 650201, China
| |
Collapse
|
11
|
Li H, Shen J, Ma S, Zhao F, Zhao W, Chen F, Fu Y, Li B, Cheng J, Deng Y. TGF-β1 suppresses de novo cholesterol biosynthesis in granulosa-lutein cells by down-regulating DHCR24 expression via the GSK-3β/EZH2/H3K27me3 signaling pathway. Int J Biol Macromol 2022; 224:1118-1128. [DOI: 10.1016/j.ijbiomac.2022.10.196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022]
|
12
|
Sapuleni J, Szymanska M, Meidan R. Diverse actions of sirtuin-1 on ovulatory genes and cell death pathways in human granulosa cells. Reprod Biol Endocrinol 2022; 20:104. [PMID: 35840944 PMCID: PMC9284863 DOI: 10.1186/s12958-022-00970-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human granulosa-lutein cells (hGLCs) amply express sirtuin-1 (SIRT1), a NAD + -dependent deacetylase that is associated with various cellular functions. SIRT1 was shown to elevate cAMP on its own and additively with human chorionic gonadotropin (hCG), it is therefore interesting to examine if SIRT1 affects other essential hGLC functions. METHODS Primary hGLCs, obtained from the follicular aspirates of women undergoing IVF and SV40-transfected, immortalized hGLCs (SVOG cells), were used. Primary cells were treated with SIRT1 specific activator SRT2104, as well as hCG or their combination. Additionally, siRNA-targeting SIRT1 construct was used to silence endogenous SIRT1 in SVOG cells. PTGS2, EREG, VEGFA and FGF2 expression was determined using quantitative polymerase chain reaction (qPCR). Apoptotic and necroptotic proteins were determined by specific antibodies in western blotting. Cell viability/apoptosis was determined by the XTT and flow cytometry analyses. Data were analyzed using student t-test or Mann-Whitney U test or one-way ANOVA followed by Tukey HSD post hoc test. RESULTS In primary and immortalized hGLCs, SRT2104 significantly upregulated key ovulatory and angiogenic genes: PTGS2, EREG, FGF2 and VEGFA, these effects tended to be further augmented in the presence of hCG. Additionally, SRT2104 dose and time-dependently decreased viable cell numbers. Flow cytometry of Annexin V stained cells confirmed that SIRT1 reduced live cell numbers and increased late apoptotic and necrotic cells. Moreover, we found that SIRT1 markedly reduced anti-apoptotic BCL-XL and MCL1 protein levels and increased cleaved forms of pro-apoptotic proteins caspase-3 and PARP. SIRT1 also significantly induced necroptotic proteins RIPK1 and MLKL. RIPK1 inhibitor, necrostatin-1 mitigated SIRT1 actions on RIPK1 and MLKL but also on cleaved caspase-3 and PARP and in accordance on live and apoptotic cells, implying a role for RIPK1 in SIRT1-induced cell death. SIRT1 silencing produced inverse effects on sorted cell populations, anti-apoptotic, pro-apoptotic and necroptotic proteins, corroborating SIRT1 activation. CONCLUSIONS These findings reveal that in hGLCs, SIRT1 enhances the expression of ovulatory and angiogenic genes while eventually advancing cell death pathways. Interestingly, these seemingly contradictory events may have occurred in a cAMP-dependent manner.
Collapse
Affiliation(s)
- Jackson Sapuleni
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 761001, Rehovot, Israel
| | - Magdalena Szymanska
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 761001, Rehovot, Israel
- Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Rina Meidan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 761001, Rehovot, Israel.
| |
Collapse
|
13
|
Luo X, Chang HM, Yi Y, Sun Y, Leung PCK. Bone morphogenetic protein 2 inhibits growth differentiation factor 8-induced cell signaling via upregulation of gremlin2 expression in human granulosa-lutein cells. Reprod Biol Endocrinol 2021; 19:173. [PMID: 34838049 PMCID: PMC8626944 DOI: 10.1186/s12958-021-00854-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/16/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bone morphogenetic protein 2 (BMP2), growth differentiation factor 8 (GDF8) and their functional receptors are expressed in human ovarian follicles, and these two intrafollicular factors play essential roles in regulating follicle development and luteal function. As BMP antagonists, gremlin1 (GREM1) and gremlin2 (GREM2) suppress BMP signaling through blockage of ligand-receptor binding. However, whether BMP2 regulates the expression of GREM1 and GREM2 in follicular development remains to be determined. METHODS In the present study, we investigated the effect of BMP2 on the expression of GREM1 and GREM2 and the underlying mechanisms in human granulosa-lutein (hGL) cells. An established immortalized human granulosa cell line (SVOG) and primary hGL cells were used as study models. The expression of GREM1 and GREM2 were examined following cell incubation with BMP2 at different concentrations and time courses. The TGF-β type I inhibitors (dorsomorphin, DMH-1 and SB431542) and small interfering RNAs targeting ALK2, ALK3, SMAD2/3, SMAD1/5/8 and SMAD4 were used to investigate the involvement of the SMAD-dependent pathway. RESULTS Our results showed that BMP2 significantly increased the expression of GREM2 (but not GREM1) in a dose- and time-dependent manner. Using a dual inhibition approach combining kinase inhibitors and siRNA-mediated knockdown, we found that the BMP2-induced upregulation of GREM2 expression was mediated by the ALK2/3-SMAD1/5-SMAD4 signaling pathway. Moreover, we demonstrated that BMP2 pretreatment significantly attenuated the GDF8-induced phosphorylation of SMAD2 and SMAD3, and this suppressive effect was reversed by knocking down GREM2 expression. CONCLUSIONS Our findings provide new insight into the molecular mechanisms by which BMP2 modulates the cellular activity induced by GDF8 through the upregulated expression of their antagonist (GREM2).
Collapse
Affiliation(s)
- Xiaoyan Luo
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, 450052, Henan, China
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Room 317, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, Zhengzhou, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Room 317, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
| | - Yuyin Yi
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Room 317, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, 450052, Henan, China.
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, Zhengzhou, China.
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Room 317, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| |
Collapse
|
14
|
Abstract
The reproductive lifespan of female mammals is limited and ultimately depends on the production of a sufficient number of high quality oocytes from a pool of non-growing primordial follicles that are set aside during embryonic and perinatal development. Recent studies show multiple signaling pathways are responsible for maintaining primordial follicle arrest and regulation of activation. Identification of these pathways and their regulatory mechanisms is essential for developing novel treatments for female infertility, improving existing in vitro fertilization techniques, and more recently, restoring the function of cryopreserved ovarian tissue. This review focuses on recent developments in transforming growth factor beta (TGFβ) family signaling in ovarian follicle development and its potential application to therapeutic design. Mouse models have been an essential tool for discovering genes critical for fertility, and recent advancements in human organ culture have additionally allowed for the translation of murine discoveries into human research and clinical settings.
Collapse
|
15
|
Wan H, Huang X, Cong P, He M, Chen A, Wu T, Dai D, Li W, Gao X, Tian L, Liang H, Xiong L. Identification of Hub Genes and Pathways Associated With Idiopathic Pulmonary Fibrosis via Bioinformatics Analysis. Front Mol Biosci 2021; 8:711239. [PMID: 34476240 PMCID: PMC8406749 DOI: 10.3389/fmolb.2021.711239] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/02/2021] [Indexed: 12/29/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease whose etiology remains unknown. The purpose of this study was to explore hub genes and pathways related to IPF development and prognosis. Multiple gene expression datasets were downloaded from the Gene Expression Omnibus database. Weighted correlation network analysis (WGCNA) was performed and differentially expressed genes (DEGs) identified to investigate Hub modules and genes correlated with IPF. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and protein-protein interaction (PPI) network analysis were performed on selected key genes. In the PPI network and cytoHubba plugin, 11 hub genes were identified, including ASPN, CDH2, COL1A1, COL1A2, COL3A1, COL14A1, CTSK, MMP1, MMP7, POSTN, and SPP1. Correlation between hub genes was displayed and validated. Expression levels of hub genes were verified using quantitative real-time PCR (qRT-PCR). Dysregulated expression of these genes and their crosstalk might impact the development of IPF through modulating IPF-related biological processes and signaling pathways. Among these genes, expression levels of COL1A1, COL3A1, CTSK, MMP1, MMP7, POSTN, and SPP1 were positively correlated with IPF prognosis. The present study provides further insights into individualized treatment and prognosis for IPF.
Collapse
Affiliation(s)
- Hanxi Wan
- Department of Anesthesiology and Perioperative Medicine, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Xinwei Huang
- Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Peilin Cong
- Department of Anesthesiology and Perioperative Medicine, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Mengfan He
- Department of Anesthesiology and Perioperative Medicine, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Aiwen Chen
- Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Tingmei Wu
- Department of Anesthesiology and Perioperative Medicine, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Danqing Dai
- Department of Anesthesiology and Perioperative Medicine, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Wanrong Li
- Department of Anesthesiology and Perioperative Medicine, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Xiaofei Gao
- Department of Anesthesiology and Perioperative Medicine, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Li Tian
- Department of Anesthesiology and Perioperative Medicine, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
| | - Huazheng Liang
- Department of Anesthesiology and Perioperative Medicine, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Lize Xiong
- Department of Anesthesiology and Perioperative Medicine, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, School of Medcine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China.,Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
| |
Collapse
|