1
|
Du X, Liu R, Jiang Z, Zhang C, Yang Z, Hu S, Zhang Z. Chondrocyte lysates activate NLRP3 inflammasome-induced pyroptosis in synovial fibroblasts to exacerbate knee synovitis by downregulating caveolin-1. Arthritis Res Ther 2025; 27:104. [PMID: 40375346 PMCID: PMC12083164 DOI: 10.1186/s13075-025-03573-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Synovitis, among the most common signs of early-stage osteoarthritis (OA), is mainly mediated by fibroblast-like synoviocytes (FLSs). Cartilage destruction creates chondrocyte lysates (CLs) that activate synovial inflammation. A comprehensive understanding of chondrocyte-FLS communication might offer novel, specific therapeutic targets for treating synovitis and OA. Hence, we sought to uncover the specific role of CLs in OA-FLSs and synovitis. METHODS Isolated CLs were cocultured with FLSs to test whether they could stimulate synovial inflammation. A model of medial meniscus destabilization was prepared in C57BL/6 mice and NLRP3 knockout mice, and adeno-associated virus overexpressing Caveolin-1 (CAV1) was intra-articularly injected for 8 weeks once a week after dissection of the medial meniscus (DMM). Proteins expressed in FLSs with and without CL coculture were screened using liquid chromatography-tandem mass spectrometry to identify CL-specific regulators of NLRP3 inflammasome-mediated pyroptosis. RESULTS CLs were engulfed by FLSs, which aggravated inflammatory cytokine release and NLRP3 inflammasome-mediated FLS pyroptosis. NLRP3 expression was significantly upregulated in human OA-FLSs and FLSs cocultured with CLs, while CAV1 was downregulated. CAV1 overexpression reversed the inflammatory phenotype in FLSs and simultaneously rescued pyroptosis in CL-pre-treated FLSs. Both synovial hyperplasia and inflammatory infiltration in C57BL/6 mice with DMM surgery were alleviated after intra-articular AAV-CAV1 injection. Moreover, the CL-specific protein LIM-containing lipoma preferred partner (LPP) markedly exacerbated FLS pyroptosis and inflammation. CONCLUSIONS CLs were endocytosed by FLSs through CAV1, and the CL-specific protein LPP stimulated NLRP3 inflammasome-mediated pyroptosis and synovitis by inhibiting CAV1 expression. Our findings offer a novel therapeutic target for treating synovitis.
Collapse
Affiliation(s)
- Xue Du
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Shandong, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Ruonan Liu
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Zongrui Jiang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Chengyun Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Zhijian Yang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Shu Hu
- Department of Joint Surgery, Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.
- Department of Joint Surgery and Sports Medicine, The Third Affiliated Hospital of Southern Medical University, Shandong, China.
| | - Zhiqi Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
2
|
Mazzetto E, Bortolami A, Bovo D, Stocchero M, Mazzacan E, Napolitan A, Panzarin V, Tran MR, Zamperin G, Milani A, Fortin A, Bigolaro M, Pirillo P, Pagliari M, Zanardello C, Giordano G, Gervasi MT, Baraldi E, Terregino C, Giaquinto C, Bonfante F. Infectivity in full-term placenta of Zika viruses with different lipid profiles. Virus Res 2025; 352:199518. [PMID: 39733819 PMCID: PMC11761821 DOI: 10.1016/j.virusres.2024.199518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/31/2024]
Abstract
Among flaviviruses, Zika virus (ZIKV) is the only arbovirus officially recognized as a teratogenic agent, as a consequence of its ability to infect and cross the placental barrier causing congenital malformation in the fetus. While many studies have focused on understanding ZIKV pathogenesis during pregnancy, the viral mechanisms affecting fetal development remain largely unclear. In this study, we investigated ZIKV virulence in placental trophoblasts, using viruses with distinct lipid profiles. Firstly, we propagated a ZIKV strain belonging to the Asian lineage in either mammalian or mosquito cells, obtaining two viral stocks, which were purified and analyzed to determine their genetic and lipid composition. Successively, we assessed the infectivity of the two stocks in placental cells using both immortalized cell lines and explants. We found that the two viral stocks displayed identical consensus sequences with homogeneous quasispecies composition. However, the lipid composition of their envelope significantly varied depending on the cell of origin, with the mammalian-derived viral stock characterized by a higher content of phosphatidylcholines compared to the virions originating from mosquito cells. Notably, ZIKV stock derived from mammalian cells showed a higher infectivity in immortalized villous trophoblasts and full-term placental explants of human origin. This increased infectivity was linked to enhanced fusion efficiency during the viral uncoating phase in trophoblast cells, as demonstrated using a lipophilic probe. Collectively, our data suggest a potential role of viral lipids as determinants of ZIKV infectivity in full-term placenta, underscoring the importance of lipidomic research in virology.
Collapse
Affiliation(s)
- Eva Mazzetto
- Department of Women's and Children's Health, Padua University, Padova (PD), Italy; Department of Virology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy.
| | - Alessio Bortolami
- Department of Virology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Davide Bovo
- Laboratory of Mass Spectrometry and Metabolomics, Department of Women's and Children's Health, Padua University, Padova (PD), Italy
| | - Matteo Stocchero
- Department of Women's and Children's Health, Padua University, Padova (PD), Italy; Laboratory of Mass Spectrometry and Metabolomics, Department of Women's and Children's Health, Padua University, Padova (PD), Italy
| | - Elisa Mazzacan
- Department of Virology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Alessandra Napolitan
- Department of Virology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Valentina Panzarin
- Department of Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Maria Rosa Tran
- Gynaecology and Obstetrics Unit, Department of Women's and Children's Health, Padua University, Padova (PD), Italy
| | - Gianpiero Zamperin
- Department of Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Adelaide Milani
- Department of Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Andrea Fortin
- Department of Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Michela Bigolaro
- Department of Diagnostic Services, Histopathology, Parasitology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Paola Pirillo
- Department of Women's and Children's Health, Padua University, Padova (PD), Italy; Laboratory of Mass Spectrometry and Metabolomics, Department of Women's and Children's Health, Padua University, Padova (PD), Italy
| | - Matteo Pagliari
- Department of Women's and Children's Health, Padua University, Padova (PD), Italy; Department of Virology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Claudia Zanardello
- Department of Diagnostic Services, Histopathology, Parasitology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Giuseppe Giordano
- Department of Women's and Children's Health, Padua University, Padova (PD), Italy; Laboratory of Mass Spectrometry and Metabolomics, Department of Women's and Children's Health, Padua University, Padova (PD), Italy
| | - Maria Teresa Gervasi
- Gynaecology and Obstetrics Unit, Department of Women's and Children's Health, Padua University, Padova (PD), Italy
| | - Eugenio Baraldi
- Department of Women's and Children's Health, Padua University, Padova (PD), Italy; Laboratory of Mass Spectrometry and Metabolomics, Department of Women's and Children's Health, Padua University, Padova (PD), Italy
| | - Calogero Terregino
- Department of Virology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| | - Carlo Giaquinto
- Department of Women's and Children's Health, Padua University, Padova (PD), Italy
| | - Francesco Bonfante
- Department of Virology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (PD), Italy
| |
Collapse
|
3
|
Wu S, Guo N, Xu H, Li Y, Sun T, Jiang X, Fu D, You T, Diao S, Huang Y, Hu C. Caveolin-1 ameliorates hepatic injury in non-alcoholic fatty liver disease by inhibiting ferroptosis via the NOX4/ROS/GPX4 pathway. Biochem Pharmacol 2024; 230:116594. [PMID: 39490677 DOI: 10.1016/j.bcp.2024.116594] [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: 05/04/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease globally, with a complex and contentious pathogenesis. Caveolin-1 (CAV1) is an important regulator of liver function and can mitigate liver injury by scavenging reactive oxygen species (ROS). Evidence suggests that NOX4 is a source of ROS production, that oxidative stress and ferroptosis are closely related, and that both are involved in the onset and progression of NAFLD. However, whether CAV1 attenuates liver injury in NAFLD caused by high-fat diet via the NOX4/ROS/GPX4 pathway remains unclear. An in vivo fatty liver model was established by feeding mice with a high-fat diet for 16 weeks. In addition, an in vitro fatty liver model was established by incubating AML-12 cells with free fatty acids for 24 h using an in vitro culture method. In our study, it was observed that a high-fat diet induces mitochondrial damage and worsens oxidative stress in NAFLD. This diet also hinders GPX4 expression, leading to an escalation of ferroptosis and lipid accumulation. To counteract these effects, intraperitoneal administration of CSD peptide in mice attenuated the high-fat diet-induced liver mitochondrial damage and ferroptosis. Likewise, overexpression of CAV1 resulted in an increase in GPX4 expression and a reduction in levels of ROS-mediated iron metamorphosis, thus mitigating the progression of the disease. However, the effects of CAV1 on GPX4-mediated ferroptosis and lipid deposition could be reversed by CAV1 small interfering RNA (SiRNA). Finally, NOX4 inhibitor (GLX351322) treatment increased CAV1 siRNA-mediated GPX4 expression and decreased the level of ROS-mediated ferroptosis. These findings suggest a potential mechanism underlying the protective role of CAV1 against high-fat diet-induced hepatotoxicity in NAFLD, shedding new light on the interplay between CAV1, GPX4, and ferroptosis in liver pathology.
Collapse
Affiliation(s)
- Shuai Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Ning Guo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Hanlin Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Tianyin Sun
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Xiangfu Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Dongdong Fu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Tingyu You
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Shaoxi Diao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Chengmu Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
4
|
Shi J, Wei L. ROCK1 deficiency preserves caveolar compartmentalization of signaling molecules and cell membrane integrity. FASEB Bioadv 2024; 6:85-102. [PMID: 38463696 PMCID: PMC10918988 DOI: 10.1096/fba.2024-00015] [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: 01/19/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
In this study, we investigated the roles of ROCK1 in regulating structural and functional features of caveolae located at the cell membrane of cardiomyocytes, adipocytes, and mouse embryonic fibroblasts (MEFs) as well as related physiopathological effects. Caveolae are small bulb-shaped cell membrane invaginations, and their roles have been associated with disease conditions. One of the unique features of caveolae is that they are physically linked to the actin cytoskeleton that is well known to be regulated by RhoA/ROCKs pathway. In cardiomyocytes, we observed that ROCK1 deficiency is coincident with an increased caveolar density, clusters, and caveolar proteins including caveolin-1 and -3. In the mouse cardiomyopathy model with transgenic overexpressing Gαq in myocardium, we demonstrated the reduced caveolar density at cell membrane and reduced caveolar protein contents. Interestingly, coexisting ROCK1 deficiency in cardiomyocytes can rescue these defects and preserve caveolar compartmentalization of β-adrenergic signaling molecules including β1-adrenergic receptor and type V/VI adenylyl cyclase. In cardiomyocytes and adipocytes, we detected that ROCK1 deficiency increased insulin signaling with increased insulin receptor activation in caveolae. In MEFs, we identified that ROCK1 deficiency increased caveolar and total levels of caveolin-1 and cell membrane repair ability after mechanical or chemical disruptions. Together, these results demonstrate that ROCK1 can regulate caveolae plasticity and multiple functions including compartmentalization of signaling molecules and cell membrane repair following membrane disruption by mechanical force and oxidative damage. These findings provide possible molecular insights into the beneficial effects of ROCK1 deletion/inhibition in cardiomyocytes, adipocytes, and MEFs under certain diseased conditions.
Collapse
Affiliation(s)
- Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of MedicineIndiana UniversityIndianapolisIndianaUSA
| |
Collapse
|
5
|
Jiang X, Li Y, Fu D, You T, Wu S, Xin J, Wen J, Huang Y, Hu C. Caveolin-1 ameliorates acetaminophen-aggravated inflammatory damage and lipid deposition in non-alcoholic fatty liver disease via the ROS/TXNIP/NLRP3 pathway. Int Immunopharmacol 2023; 114:109558. [PMID: 36700765 DOI: 10.1016/j.intimp.2022.109558] [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: 10/11/2022] [Revised: 11/24/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
The overuse of acetaminophen (APAP) may cause more severe hepatotoxicity in patients with non-alcoholic fatty liver disease (NAFLD). Caveolin-1 (CAV1), is an essential regulator of metabolic function, which can alleviate liver damage by scavenging reactive oxygen species (ROS). Evidence suggests that the NOD-like receptor family pyrin domain-containing 3 (NLRP3) -mediated pyroptosis is involved in the development of NAFLD. Moreover, thioredoxin-interactive protein (TXNIP) activation is a key event linking ROS to NLRP3 inflammasome. However, whether CAV1 alleviates APAP-aggravated hepatotoxicity in NAFLD via the ROS/TXNIP/NLRP3 pathway remains unclear. An in vivo fatty liver model was established by feeding mice a high-fat diet for 56 days. Additionally, using in vitro approach, AML-12 cells were incubated with free fatty acids for 48 h and APAP was added during the last 24 h. We found that the overuse of APAP in NAFLD not only induced oxidative stress, but also increased TXNIP expression, NLRP3-mediated pyroptosis, and lipid deposition. In addition to inhibiting ROS generation and lipid deposition, overexpression of CAV1 reduced the elevated levels of TXNIP expression and NLRP3-mediated pyroptosis. However, the effect of CAV1 on TXNIP expression, NLRP3-mediated pyroptosis, and lipid deposition was reversed by CAV1 small interfering RNA (siRNA) intervention. Finally, N-acetyl cysteine (NAC) treatment reduced CAV1 siRNA-mediated changes in TXNIP expression and NLRP3-mediated pyroptosis levels. These results demonstrate that the inhibitory effect of CAV1 on NLRP3-mediated pyroptosis may be mediated through the ROS/TXNIP axis. Moreover, the current study provides novel mechanistic insights into the protective effects of CAV1 on APAP-aggravated hepatotoxicity in NAFLD.
Collapse
Affiliation(s)
- Xiangfu Jiang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Yu Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Dongdong Fu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Tingyu You
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Shuai Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Jiao Xin
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Jiagen Wen
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Yan Huang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Chengmu Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
6
|
Expression of S100 proteins is associated with HBV intrauterine transmission. Arch Gynecol Obstet 2020; 302:1389-1399. [PMID: 32856138 DOI: 10.1007/s00404-020-05753-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/15/2020] [Indexed: 12/26/2022]
Abstract
PURPOSE The mechanisms underlying HBV intrauterine transmission remain unknown. In this study, we explored the mechanism of HBV intrauterine transmission by iTRAQ proteomics analysis. METHODS iTRAQ technology was applied to perform comparative proteomics studies on six HBV+/+ neonates and six HBV+/- neonates whose mothers and fathers were HBsAg positive and paternal HBsAg negative, respectively. The data obtained from the mass spectrometer were analyzed using MASCOT ( https://matrixscience.com ) to qualitatively and quantitatively compare the differentially expressed proteins in the two groups. Gene Ontology and KEGG pathway analyses were performed to analyze the differentially expressed proteins. The expressions of HBV intrauterine transmission-related proteins in serum samples and corresponding placental tissues were further verified by immunohistochemistry and Western Blot. Then, the human trophoblast cell line (Swan71) infected with HBV was used to analyze the potential mechanisms of HBV intrauterine transmission under the mediation of differential proteins. RESULTS A total of 35 differentially expressed proteins, including 17 up-regulated proteins and 18 down-regulated proteins, were identified by comparing serum protein expression levels in HBV+/+ and HBV+/- neonates. The differentially expressed proteins were mainly related to RAGE receptor binding, NF-kappa B transcription factor activity, innate immune response, defense response to bacterium, and the signaling pathway in pathogenic microorganism infection. The expressions of S100A8/9/12 in HBV+/+ maternal placenta tissue were significantly increased. The expressions of S100A8/9/12 proteins in Swan71 cells were significantly increased after HBV infection. CONCLUSION High expression of S100 proteins may be associated with the intrauterine-transplacental transmission of HBV.
Collapse
|
7
|
Ducat A, Couderc B, Bouter A, Biquard L, Aouache R, Passet B, Doridot L, Cohen MB, Ribaux P, Apicella C, Gaillard I, Palfray S, Chen Y, Vargas A, Julé A, Frelin L, Cocquet J, San Martin CR, Jacques S, Busato F, Tost J, Méhats C, Laissue P, Vilotte JL, Miralles F, Vaiman D. Molecular Mechanisms of Trophoblast Dysfunction Mediated by Imbalance between STOX1 Isoforms. iScience 2020; 23:101086. [PMID: 32371375 PMCID: PMC7200942 DOI: 10.1016/j.isci.2020.101086] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/16/2020] [Accepted: 04/15/2020] [Indexed: 12/16/2022] Open
Abstract
STOX1 is a transcription factor involved in preeclampsia and Alzheimer disease. We show that the knock-down of the gene induces rather mild effect on gene expression in trophoblast cell lines (BeWo). We identified binding sites of STOX1 shared by the two major isoforms, STOX1A and STOX1B. Profiling gene expression of cells overexpressing either STOX1A or STOX1B, we identified genes downregulated by both isoforms, with a STOX1 binding site in their promoters. Among those, STOX1-induced Annexin A1 downregulation led to abolished membrane repair in BeWo cells. By contrast, overexpression of STOX1A or B has opposite effects on trophoblast fusion (acceleration and inhibition, respectively) accompanied by syncytin genes deregulation. Also, STOX1A overexpression led to abnormal regulation of oxidative and nitrosative stress. In sum, our work shows that STOX1 isoform imbalance is a cause of gene expression deregulation in the trophoblast, possibly leading to placental dysfunction and preeclampsia.
Collapse
Affiliation(s)
- Aurélien Ducat
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Betty Couderc
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Anthony Bouter
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, IPB, 33600 Pessac, France
| | - Louise Biquard
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Rajaa Aouache
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Bruno Passet
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Ludivine Doridot
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Marie-Benoîte Cohen
- Department of Gynecology Obstetrics, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Pascale Ribaux
- Department of Gynecology Obstetrics, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Clara Apicella
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Irène Gaillard
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Sophia Palfray
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Yulian Chen
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Alexandra Vargas
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Amélie Julé
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Léo Frelin
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, IPB, 33600 Pessac, France
| | - Julie Cocquet
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Camino Ruano San Martin
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Sébastien Jacques
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Florence Busato
- Laboratory for Epigenetics and Environment, Institut de Biologie François Jacob, Commissariat àl'Energie Atomique, Evry 91057, France
| | - Jorg Tost
- Laboratory for Epigenetics and Environment, Institut de Biologie François Jacob, Commissariat àl'Energie Atomique, Evry 91057, France
| | - Céline Méhats
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Paul Laissue
- Biopas Laboratoires, BIOPAS GROUP, Bogotá, Colombia
| | - Jean-Luc Vilotte
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Francisco Miralles
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France
| | - Daniel Vaiman
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université Paris Descartes, Paris 75014, France.
| |
Collapse
|
8
|
Yu B, Sladojevic N, Blair JE, Liao JK. Targeting Rho-associated coiled-coil forming protein kinase (ROCK) in cardiovascular fibrosis and stiffening. Expert Opin Ther Targets 2020; 24:47-62. [PMID: 31906742 PMCID: PMC7662835 DOI: 10.1080/14728222.2020.1712593] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 01/04/2020] [Indexed: 02/07/2023]
Abstract
Introduction: Pathological cardiac fibrosis, through excessive extracellular matrix protein deposition from fibroblasts and pro-fibrotic immune responses and vascular stiffening is associated with most forms of cardiovascular disease. Pathological cardiac fibrosis and stiffening can lead to heart failure and arrythmias and vascular stiffening may lead to hypertension. ROCK, a serine/threonine kinase downstream of the Rho-family of GTPases, may regulate many pro-fibrotic and pro-stiffening signaling pathways in numerous cell types.Areas covered: This article outlines the molecular mechanisms by which ROCK in fibroblasts, T helper cells, endothelial cells, vascular smooth muscle cells, and macrophages mediate fibrosis and stiffening. We speculate on how ROCK could be targeted to inhibit cardiovascular fibrosis and stiffening.Expert opinion: Critical gaps in knowledge must be addressed if ROCK inhibitors are to be used in the clinic. Numerous studies indicate that each ROCK isoform may play differential roles in regulating fibrosis and may have opposing roles in specific tissues. Future work needs to highlight the isoform- and tissue-specific contributions of ROCK in fibrosis, and how isoform-specific ROCK inhibitors in murine models and in clinical trials affect the pathophysiology of cardiac fibrosis and stiffening. This could progress knowledge regarding new treatments for heart failure, arrythmias and hypertension and the repair processes after myocardial infarction.
Collapse
Affiliation(s)
- Brian Yu
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Nikola Sladojevic
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - John E Blair
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - James K Liao
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
9
|
Reppetti J, Reca A, Seyahian EA, Medina Y, Martínez N, Szpilbarg N, Damiano AE. Intact caveolae are required for proper extravillous trophoblast migration and differentiation. J Cell Physiol 2019; 235:3382-3392. [DOI: 10.1002/jcp.29226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/23/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Julieta Reppetti
- Laboratorio de Biología de la Reproducción, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)‐CONICET‐ Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
| | - Alejandra Reca
- Laboratorio de Biología de la Reproducción, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)‐CONICET‐ Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
| | - E. Abril Seyahian
- Laboratorio de Biología de la Reproducción, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)‐CONICET‐ Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
| | - Yollyseth Medina
- Laboratorio de Biología de la Reproducción, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)‐CONICET‐ Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
| | - Nora Martínez
- Laboratorio de Biología de la Reproducción, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)‐CONICET‐ Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
| | - Natalia Szpilbarg
- Laboratorio de Biología de la Reproducción, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)‐CONICET‐ Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
| | - Alicia E. Damiano
- Laboratorio de Biología de la Reproducción, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)‐CONICET‐ Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
- Cátedra de Biología Celular y Molecular, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica Universidad de Buenos Aires Buenos Aires Argentina
| |
Collapse
|
10
|
Chung CL, Wang SW, Sun WC, Shu CW, Kao YC, Shiao MS, Chen CL. Sorafenib suppresses TGF-β responses by inducing caveolae/lipid raft-mediated internalization/degradation of cell-surface type II TGF-β receptors: Implications in development of effective adjunctive therapy for hepatocellular carcinoma. Biochem Pharmacol 2018; 154:39-53. [PMID: 29678520 DOI: 10.1016/j.bcp.2018.04.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/13/2018] [Indexed: 12/31/2022]
Abstract
Sorafenib is the only FDA approved drug for the treatment of advanced hepatocellular carcinoma (HCC) and other malignancies. Studies indicate that TGF-β signalling is associated with tumour progression in HCC. Autocrine and paracrine TGF-β promotes tumour growth and malignancy by inducing epithelial-mesenchymal transition (EMT). Sorafenib is believed to antagonize tumour progression by inhibiting TGF-β-induced EMT. It improves survival of patients but HCC later develops resistance and relapses. The underlying mechanism of resistance is unknown. Understanding of the molecular mechanism of sorafenib inhibition of TGF-β-induced signalling or responses in HCC may lead to development of adjunctive effective therapy for HCC. In this study, we demonstrate that sorafenib suppresses TGF-β responsiveness in hepatoma cells, hepatocytes, and animal liver, mainly by downregulating cell-surface type II TGF-β receptors (TβRII) localized in caveolae/lipid rafts and non-lipid raft microdomains via caveolae/lipid rafts-mediated internalization and degradation. Furthermore, sorafenib-induced downregulation and degradation of cell-surface TβRII is prevented by simultaneous treatment with a caveolae disruptor or lysosomal inhibitors. On the other hand, sorafenib only downregulates cell-surface TβRII localized in caveolae/lipid rafts but not localized in non-lipid raft microdomains in hepatic stellate cells. These results suggest that sorafenib inhibits TGF-β signalling mainly by inducing caveolae/lipid raft-mediated internalization and degradation of cell-surface TβR-II in target cells. They may also imply that treatment with agents which promote formation of caveolae/lipid rafts, TGF-β receptor kinase inhibitors (e.g., LY2157299) or TGF-β peptide antagonists (by liver-targeting delivery) may be considered as effective adjunct therapy with sorafenib for HCC.
Collapse
Affiliation(s)
- Chih-Ling Chung
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC
| | - Shih-Wei Wang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC
| | - Wei-Chih Sun
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81326, Taiwan, ROC
| | - Chih-Wen Shu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81326, Taiwan, ROC
| | - Yu-Chen Kao
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC
| | - Meng-Shin Shiao
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan, ROC.
| |
Collapse
|
11
|
Ushiyama A, Tajima A, Ishikawa N, Asano A. Characterization of the functions and proteomes associated with membrane rafts in chicken sperm. PLoS One 2017; 12:e0186482. [PMID: 29095853 PMCID: PMC5667776 DOI: 10.1371/journal.pone.0186482] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/02/2017] [Indexed: 01/23/2023] Open
Abstract
Cellular membranes are heterogeneous, and this has a great impact on cellular function. Despite the central role of membrane functions in multiple cellular processes in sperm, their molecular mechanisms are poorly understood. Membrane rafts are specific membrane domains enriched in cholesterol, ganglioside GM1, and functional proteins, and they are involved in the regulation of a variety of cellular functions. Studies of the functional characterization of membrane rafts in mammalian sperm have demonstrated roles in sperm-egg binding and the acrosomal reaction. Recently, our biochemical and cell biological studies showed that membrane rafts are present and might play functional roles in chicken sperm. In this study, we isolated membrane rafts from chicken sperm as a detergent-resistant membranes (DRM) floating on a density gradient in the presence of 1% Triton X-100, and characterized the function and proteomes associated with these domains. Biochemical comparison of the DRM between fresh and cryopreserved sperm demonstrated that cryopreservation induces cholesterol loss specifically from membrane rafts, indicating the functional connection with reduced post-thaw fertility in chicken sperm. Furthermore, using an avidin-biotin system, we found that sperm DRM is highly enriched in a 60 KDa single protein able to bind to the inner perivitelline layer. To identify possible roles of membrane rafts, quantitative proteomics, combined with a stable isotope dimethyl labeling approach, identified 82 proteins exclusively or relatively more associated with membrane rafts. Our results demonstrate the functional distinctions between membrane domains and provide compelling evidence that membrane rafts are involved in various cellular pathways inherent to chicken sperm.
Collapse
Affiliation(s)
- Ai Ushiyama
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan
| | - Atsushi Tajima
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan
| | - Naoto Ishikawa
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan
| | - Atsushi Asano
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
12
|
Levi L, Castro-Parodi M, Martínez N, Piehl LL, Rubín De Celis E, Herlax V, Mate S, Farina M, Damiano AE. The unfavorable lipid environment reduced caveolin-1 expression in apical membranes from human preeclamptic placentas. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2171-2180. [DOI: 10.1016/j.bbamem.2016.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/27/2016] [Accepted: 06/17/2016] [Indexed: 01/29/2023]
|
13
|
Chen TW, Liu HW, Liou YJ, Lee JH, Lin CH. Over-expression of stomatin causes syncytium formation in nonfusogenic JEG-3 choriocarcinoma placental cells. Cell Biol Int 2016; 40:926-33. [PMID: 27306251 DOI: 10.1002/cbin.10636] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/11/2016] [Indexed: 11/09/2022]
Abstract
Placental trophoblast differentiation involves the continuous fusion of mononuclear cytotrophoblasts. However, except for syncytin, little is known about the detailed mechanisms underlying trophoblast fusion. A previous study indicated that lipid rafts play an important role in HTLV-1 syncytium formation. To identify proteins that may be involved in placental trophoblast differentiation, we examined stomatin, an important lipid-raft protein that localizes to detergent-resistant membrane domains. The syncytium and human chorionic gonadotropin (β-hCG; a marker of placental trophoblast differentiation) were visualized by immunofluorescence staining. We found that overexpression of stomatin in the nonfusogenic JEG-3 cell line caused syncytium formation and increased the fusion index of cells. Treating these cells with N(6) ,2'-O-dibutyryladenosine 3',5'-cyclic monophosphate further increased cell fusion by stomatin. β-hCG was found in a few JEG-3 cells overexpressing stomatin at 48 h, and its levels increased dramatically at 72 h along with the formation of the multinuclear syncytium. RNA interference was used to decrease stomatin expression in BeWo cells, a fusogenic human choriocarcinoma cell line. After knockdown for 72 h, stomatin levels decreased by almost 95%. The fusion indexes of control and stomatin-knockdown cells at 72 h were 9.4 and 6.5%, respectively. Our data indicated that stomatin could trigger syncytium formation and upregulate β-hCG for cell fusion in nonfusogenic JEG-3 cells. Downregulation of stomatin slightly inhibited the fusion index of fusogenic BeWo cells. Thus, these data suggested that stomatin plays an important role in trophoblast differentiation.
Collapse
Affiliation(s)
- Tung-Wei Chen
- Institute of Biophotonics, National Yang-Ming University, No. 155, Sec. 2, Linong, Street, Taipei, Taiwan, 112,, ROC
| | - Hong-Wen Liu
- Institute of Microbiology and Immunology, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei, Taiwan, 112,, ROC
| | - Yi-Jia Liou
- Institute of Microbiology and Immunology, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei, Taiwan, 112,, ROC
| | - Jui-Hao Lee
- Institute of Microbiology and Immunology, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei, Taiwan, 112,, ROC.,Institute of Biochemistry, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei, Taiwan, 112,, ROC.,Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, No. 155, Sec. 2, Linong Street, Taipei, Taiwan, 112,, ROC
| | - Chi-Hung Lin
- Institute of Biophotonics, National Yang-Ming University, No. 155, Sec. 2, Linong, Street, Taipei, Taiwan, 112,, ROC.,Institute of Microbiology and Immunology, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei, Taiwan, 112,, ROC.,Department of Health, New Taipei City Government, Taipei, Taiwan
| |
Collapse
|
14
|
RhoB/ROCK mediates oxygen–glucose deprivation-stimulated syncytiotrophoblast microparticle shedding in preeclampsia. Cell Tissue Res 2016; 366:411-425. [DOI: 10.1007/s00441-016-2436-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 05/12/2016] [Indexed: 02/06/2023]
|
15
|
Huang SS, Chen CL, Huang FW, Hou WH, Huang JS. DMSO Enhances TGF-β Activity by Recruiting the Type II TGF-β Receptor From Intracellular Vesicles to the Plasma Membrane. J Cell Biochem 2016; 117:1568-79. [PMID: 26587792 DOI: 10.1002/jcb.25448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/18/2015] [Indexed: 01/03/2023]
Abstract
Dimethyl sulfoxide (DMSO) is used to treat many diseases/symptoms. The molecular basis of the pharmacological actions of DMSO has been unclear. We hypothesized that DMSO exerts some of these actions by enhancing TGF-β activity. Here we show that DMSO enhances TGF-β activity by ∼3-4-fold in Mv1Lu and NMuMG cells expressing Smad-dependent luciferase reporters. In Mv1Lu cells, DMSO enhances TGF-β-stimulated expression of P-Smad2 and PAI-1. It increases cell-surface expression of TGF-β receptors (TβR-I and/or TβR-II) by ∼3-4-fold without altering their cellular levels as determined by (125) I-labeled TGF-β-cross-linking/Western blot analysis, suggesting the presence of large intracellular pools in these cells. Sucrose density gradient ultracentrifugation/Western blot analysis reveals that DMSO induces recruitment of TβR-II (but not TβR-I) from its intracellular pool to plasma-membrane microdomains. It induces more recruitment of TβR-II to non-lipid raft microdomains than to lipid rafts/caveolae. Mv1Lu cells transiently transfected with TβR-II-HA plasmid were treated with DMSO and analyzed by indirect immunofluoresence staining using anti-HA antibody. In these cells, TβR-II-HA is present as a vesicle-like network in the cytoplasm as well as in the plasma membrane. DMSO causes depletion of TβR-II-HA-containing vesicles from the cytoplasm and co-localization of TβR-II-HA and cveolin-1 at the plasma membrane. These results suggest that DMSO, a fusogenic substance, enhances TGF-β activity presumably by inducing fusion of cytoplasmic vesicles (containing TβR-II) and the plasma membrane, resulting in increased localization of TβR-II to non-lipid raft microdomains where canonical signaling occurs. Fusogenic activity of DMSO may play a pivotal role in its pharmacological actions involving membrane proteins with large cytoplasmic pools. J. Cell. Biochem. 117: 1568-1579, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-sen University and Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 804, Taiwan
| | - Franklin W Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston and Harvard Medical School, Boston, Massachusetts, 02115
| | - Wei-Hsien Hou
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, 1100 S. Grand Boulevard, St. Louis, Missouri, 63104
| | - Jung San Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, 1100 S. Grand Boulevard, St. Louis, Missouri, 63104
| |
Collapse
|
16
|
Huang SS, Chen CL, Huang FW, Johnson FE, Huang JS. Ethanol Enhances TGF-β Activity by Recruiting TGF-β Receptors From Intracellular Vesicles/Lipid Rafts/Caveolae to Non-Lipid Raft Microdomains. J Cell Biochem 2015; 117:860-71. [PMID: 26419316 DOI: 10.1002/jcb.25389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022]
Abstract
Regular consumption of moderate amounts of ethanol has important health benefits on atherosclerotic cardiovascular disease (ASCVD). Overindulgence can cause many diseases, particularly alcoholic liver disease (ALD). The mechanisms by which ethanol causes both beneficial and harmful effects on human health are poorly understood. Here we demonstrate that ethanol enhances TGF-β-stimulated luciferase activity with a maximum of 0.5-1% (v/v) in Mv1Lu cells stably expressing a luciferase reporter gene containing Smad2-dependent elements. In Mv1Lu cells, 0.5% ethanol increases the level of P-Smad2, a canonical TGF-β signaling sensor, by ∼ 2-3-fold. Ethanol (0.5%) increases cell-surface expression of the type II TGF-β receptor (TβR-II) by ∼ 2-3-fold from its intracellular pool, as determined by I(125) -TGF-β-cross-linking/Western blot analysis. Sucrose density gradient ultracentrifugation and indirect immunofluorescence staining analyses reveal that ethanol (0.5% and 1%) also displaces cell-surface TβR-I and TβR-II from lipid rafts/caveolae and facilitates translocation of these receptors to non-lipid raft microdomains where canonical signaling occurs. These results suggest that ethanol enhances canonical TGF-β signaling by increasing non-lipid raft microdomain localization of the TGF-β receptors. Since TGF-β plays a protective role in ASCVD but can also cause ALD, the TGF-β enhancer activity of ethanol at low and high doses appears to be responsible for both beneficial and harmful effects. Ethanol also disrupts the location of lipid raft/caveolae of other membrane proteins (e.g., neurotransmitter, growth factor/cytokine, and G protein-coupled receptors) which utilize lipid rafts/caveolae as signaling platforms. Displacement of these membrane proteins induced by ethanol may result in a variety of pathologies in nerve, heart and other tissues.
Collapse
Affiliation(s)
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung, 804, Taiwan.,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 804, Taiwan
| | - Franklin W Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, 02115.,Harvard Medical School, Boston, Massachusetts, 02115
| | - Frank E Johnson
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri, 63104
| | - Jung San Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, St. Louis, Missouri, 63104
| |
Collapse
|
17
|
Wei BR, Xu C, Rote NS. Increased resistance to apoptosis during differentiation and syncytialization of BeWo choriocarcinoma cells. ACTA ACUST UNITED AC 2012; 3:805-813. [PMID: 29623239 DOI: 10.4236/abb.2012.326100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Transition from mononuclear villous cytotrophoblast into multinuclear syncytiotrophoblast in the human placenta is accompanied by changes in apoptosis-related proteins and an apparent increased resistance to induced apoptosis. We investigated the specific nature and timing of changes in Bcl-2, Bax, p53, and caspases 3 and 8 in forskolin-treated BeWo choriocarcinoma cells, a model for villous cytotrophoblast differentiation. BeWo cells were treated with forskolin or vehicle alone for up to 72 h and evaluated at 24 h intervals for syncytialization and quantitative expression specific apoptosis-related proteins and mRNAs. Syncytialization was quantified using fluorescent staining of intercellular membranes and enumeration of the percentage of nuclei in multinucleate cells, and differential localization of apoptosis-related proteins to multinuclear or mononuclear cells was determined by quantitative immunofluorescence. Forskolin treatment for up to 72 h resulted in 80% syncytialization, increased expression of Bcl-2 protein (P < 0.01) and mRNA (P < 0.05), and significantly decreased expression of protein and mRNA for Bax, p53, and caspases 3 and 8. Syncytialized cells expressed higher levels of Bcl-2 protein concurrent with increased resistance to cisplatin-induced apoptosis. Thus, syncytialization of BeWo cells was accompanied by altered transcription of apoptotic-related proteins characteristic of increased apoptosis resistance secondary to increased expression of the anti-apoptotic protein Bcl-2 and diminish expression of pro-apoptotic proteins.
Collapse
Affiliation(s)
- Bih-Rong Wei
- Department of Reproductive Biology, Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, USA.,Department of Obstetrics and Gynecology, University Hospitals Case Medical Center, Cleveland, USA.,SAIC-Frederick, Bethesda, USA
| | - Chuan Xu
- Department of Reproductive Biology, Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, USA.,Department of Obstetrics and Gynecology, University Hospitals Case Medical Center, Cleveland, USA
| | - Neal S Rote
- Department of Reproductive Biology, Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, USA.,Department of Obstetrics and Gynecology, University Hospitals Case Medical Center, Cleveland, USA
| |
Collapse
|
18
|
Walton JR, Frey HA, Vandre DD, Kwiek JJ, Ishikawa T, Takizawa T, Robinson JM, Ackerman WE. Expression of flotillins in the human placenta: potential implications for placental transcytosis. Histochem Cell Biol 2012; 139:487-500. [PMID: 23064789 DOI: 10.1007/s00418-012-1040-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2012] [Indexed: 02/07/2023]
Abstract
A proteomics survey of human placental syncytiotrophoblast (ST) apical plasma membranes revealed peptides corresponding to flotillin-1 (FLOT1) and flotillin-2 (FLOT2). The flotillins belong to a class of lipid microdomain-associated integral membrane proteins that have been implicated in clathrin- and caveolar-independent endocytosis. In the present study, we characterized the expression of the flotillin proteins within the human placenta. FLOT1 and FLOT2 were coexpressed in placental lysates and BeWo human trophoblast cells. Immunofluorescence microscopy of first-trimester and term placentas revealed that both proteins were more prominent in villous endothelial cells and cytotrophoblasts (CTs) than the ST. Correspondingly, forskolin-induced fusion in BeWo cells resulted in a decrease in FLOT1 and FLOT2, suggesting that flotillin protein expression is reduced following trophoblast syncytialization. The flotillin proteins co-localized with a marker of fluid-phase pinocytosis, and knockdown of FLOT1 and/or FLOT2 expression resulted in decreased endocytosis of cholera toxin B subunit. We conclude that FLOT1 and FLOT2 are abundantly coexpressed in term villous placental CTs and endothelial cells, and in comparison, expression of these proteins in the ST is reduced. These findings suggest that flotillin-dependent endocytosis is unlikely to be a major pathway in the ST, but may be important in the CT and endothelium.
Collapse
Affiliation(s)
- Janelle R Walton
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Byrne DP, Dart C, Rigden DJ. Evaluating caveolin interactions: do proteins interact with the caveolin scaffolding domain through a widespread aromatic residue-rich motif? PLoS One 2012; 7:e44879. [PMID: 23028656 PMCID: PMC3444507 DOI: 10.1371/journal.pone.0044879] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 08/09/2012] [Indexed: 01/08/2023] Open
Abstract
Caveolins are coat proteins of caveolae, small flask-shaped pits of the plasma membranes of most cells. Aside from roles in caveolae formation, caveolins recruit, retain and regulate many caveolae-associated signalling molecules. Caveolin-protein interactions are commonly considered to occur between a ∼20 amino acid region within caveolin, the caveolin scaffolding domain (CSD), and an aromatic-rich caveolin binding motif (CBM) on the binding partner (фXфXXXXф, фXXXXфXXф or фXфXXXXфXXф, where ф is an aromatic and X an unspecified amino acid). The CBM resembles a typical linear motif - a short, simple sequence independently evolved many times in different proteins for a specific function. Here we exploit recent improvements in bioinformatics tools and in our understanding of linear motifs to critically examine the role of CBMs in caveolin interactions. We find that sequences conforming to the CBM occur in 30% of human proteins, but find no evidence for their statistical enrichment in the caveolin interactome. Furthermore, sequence- and structure-based considerations suggest that CBMs do not have characteristics commonly associated with true interaction motifs. Analysis of the relative solvent accessible area of putative CBMs shows that the majority of their aromatic residues are buried within the protein and are thus unlikely to interact directly with caveolin, but may instead be important for protein structural stability. Together, these findings suggest that the canonical CBM may not be a common characteristic of caveolin-target interactions and that interfaces between caveolin and targets may be more structurally diverse than presently appreciated.
Collapse
Affiliation(s)
- Dominic P. Byrne
- Institute of Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
| | - Caroline Dart
- Institute of Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
| | - Daniel J. Rigden
- Institute of Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
20
|
RhoE is regulated by cyclic AMP and promotes fusion of human BeWo choriocarcinoma cells. PLoS One 2012; 7:e30453. [PMID: 22272352 PMCID: PMC3260294 DOI: 10.1371/journal.pone.0030453] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 12/16/2011] [Indexed: 11/19/2022] Open
Abstract
Fusion of placental villous cytotrophoblasts with the overlying syncytiotrophoblast is essential for the maintenance of successful pregnancy, and disturbances in this process have been implicated in pathological conditions such as pre-eclampsia and intra-uterine growth retardation. In this study we examined the role of the Rho GTPase family member RhoE in trophoblast differentiation and fusion using the BeWo choriocarcinoma cell line, a model of villous cytotrophoblast fusion. Treatment of BeWo cells with the cell permeable cyclic AMP analogue dibutyryl cyclic AMP (dbcAMP) resulted in a strong upregulation of RhoE at 24h, coinciding with the onset of fusion. Using the protein kinase A (PKA)-specific cAMP analogue N6-phenyl-cAMP, and a specific inhibitor of PKA (14–22 amide, PKI), we found that upregulation of RhoE by cAMP was mediated through activation of PKA signalling. Silencing of RhoE expression by RNA interference resulted in a significant decrease in dbcAMP-induced fusion. However, expression of differentiation markers human chorionic gonadotrophin and placental alkaline phosphatase was unaffected by RhoE silencing. Finally, we found that RhoE upregulation by dbcAMP was significantly reduced under hypoxic conditions in which cell fusion is impaired. These results show that induction of RhoE by cAMP is mediated through PKA and promotes BeWo cell fusion but has no effect on functional differentiation, supporting evidence that these two processes may be controlled by separate or diverging pathways.
Collapse
|
21
|
Asano A, Nelson JL, Zhang S, Travis AJ. Characterization of the proteomes associating with three distinct membrane raft sub-types in murine sperm. Proteomics 2011; 10:3494-505. [PMID: 20815087 DOI: 10.1002/pmic.201000002] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mammalian sperm are transcriptionally and translationally inactive. To meet changing needs in the epididymis and female tract, they rely heavily on post-translational modifications and protein acquisition/degradation. Membrane rafts are sterol and sphingolipid-enriched micro-domains that organize and regulate various pathways. Rafts have significance in sperm by transducing the stimulus of sterol efflux into changes in intracellular signaling that confer fertilization competence. We recently characterized three biochemically distinct sub-types of sperm rafts, and now present profiles for proteins targeting to and associating with these sub-types, along with a fraction largely comprised of "non-raft" domains. Proteomics analysis using a gel-based LC-MS/MS approach identified 190 strictly validated proteins in the raft sub-types. Interestingly, many of these are known to be expressed in the epididymis, where sperm membrane composition matures. To investigate potential roles for rafts in epididymal protein acquisition, we compared the expression and localization of two different sterol-interacting proteins, apolipoprotein-A1 (apoA1) and prominin-1 (prom1) in sperm from different zones. We found that apoA1 was gradually added to the plasma membrane overlying the acrosome, whereas prom1 was not, suggesting different mechanisms for raft protein acquisition. Our results define raft-associating proteins, demonstrate functional similarities and differences among raft sub-types, and provide insights into raft-mediated epididymal protein acquisition.
Collapse
Affiliation(s)
- Atsushi Asano
- The Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | |
Collapse
|
22
|
Boettcher JP, Kirchner M, Churin Y, Kaushansky A, Pompaiah M, Thorn H, Brinkmann V, MacBeath G, Meyer TF. Tyrosine-phosphorylated caveolin-1 blocks bacterial uptake by inducing Vav2-RhoA-mediated cytoskeletal rearrangements. PLoS Biol 2010; 8. [PMID: 20808760 PMCID: PMC2927421 DOI: 10.1371/journal.pbio.1000457] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Accepted: 07/07/2010] [Indexed: 12/11/2022] Open
Abstract
During the early stages of infection, Neisseria gonorrhoeae triggers a phosphotyrosine-dependent Cav1-Vav2-RhoA signaling cascade that promotes the pathogen's extracellular state. Certain bacterial adhesins appear to promote a pathogen's extracellular lifestyle rather than its entry into host cells. However, little is known about the stimuli elicited upon such pathogen host-cell interactions. Here, we report that type IV pili (Tfp)-producing Neisseria gonorrhoeae (P+GC) induces an immediate recruitment of caveolin-1 (Cav1) in the host cell, which subsequently prevents bacterial internalization by triggering cytoskeletal rearrangements via downstream phosphotyrosine signaling. A broad and unbiased analysis of potential interaction partners for tyrosine-phosphorylated Cav1 revealed a direct interaction with the Rho-family guanine nucleotide exchange factor Vav2. Both Vav2 and its substrate, the small GTPase RhoA, were found to play a direct role in the Cav1-mediated prevention of bacterial uptake. Our findings, which have been extended to enteropathogenic Escherichia coli, highlight how Tfp-producing bacteria avoid host cell uptake. Further, our data establish a mechanistic link between Cav1 phosphorylation and pathogen-induced cytoskeleton reorganization and advance our understanding of caveolin function. Like many bacterial pathogens, successful attachment of Neisseria gonorrhoeae—the causative agent of the sexually transmitted disease gonorrhoea—to its host cells depends on specialized structures on the bacterial surface called type IV pili (Tfp). Pathogen attachment induces changes within host cells that may facilitate and promote infection. In this study, we identify some of the earliest cellular signals elicited by N. gonorrhoeae during infection, which, in this case, prevent the organism from entering the cell precociously. After attachment to host cells the bacteria form microcolonies on the cell surface. Underneath these microcolonies, so-called cortical plaques form within the host cell—these contain the cytoskeleton protein actin and a range of signaling proteins. We show that N. gonorrhoeae recruits a host cell protein called caveolin-1 to the cell membrane where the bacteria are attached; here, caveloin-1 effectively impedes uptake of the bacteria by activating a signaling cascade that involves its phosphorylation on a tyrosine residue and subsequent interactions with proteins that regulate the cytoskeleton. Thus, these proteins play a pivotal role in maintaining N. gonorrhoeae in the extracellular milieu. By extrapolating our findings to another Tfp-producing bacterium, the enteropathogenic Escherichia coli, we argue that the establishment and maintenance of this extracellular state benefits certain pathogens by giving them time to express proteins required for subsequent steps of infection.
Collapse
Affiliation(s)
- Jan Peter Boettcher
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Marieluise Kirchner
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Yuri Churin
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Alexis Kaushansky
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Malvika Pompaiah
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hans Thorn
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Volker Brinkmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Gavin MacBeath
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Thomas F. Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
- * E-mail:
| |
Collapse
|
23
|
Collett GP, Linton EA, Redman CWG, Sargent IL. Downregulation of caveolin-1 enhances fusion of human BeWo choriocarcinoma cells. PLoS One 2010; 5:e10529. [PMID: 20463894 PMCID: PMC2865536 DOI: 10.1371/journal.pone.0010529] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 04/13/2010] [Indexed: 12/11/2022] Open
Abstract
Background Fusion of placental villous cytotrophoblasts with the overlying syncytiotrophoblast is essential for the maintenance of successful pregnancy, and disturbances in this process have been implicated in pathological conditions such as pre-eclampsia and intra-uterine growth retardation. Caveolin-1 has been shown to be expressed in human villous cytotrophoblast and to be downregulated during fusion into syncytiotrophoblast but it is unclear whether it plays a role in this process. Methodology/Principal Findings We used RNA interference to determine whether caveolin-1 plays a role in differentiation and fusion in the BeWo choriocarcinoma cell line, a model of villous cytotrophoblast fusion. Assessment of cell fusion by desmosomal protein immunostaining revealed that cells transfected with caveolin-1 siRNA showed significantly enhanced fusion in response to treatment with dibutyryl cyclic AMP compared with cells transfected with a non-silencing control. Furthermore, caveolin-1 knockdown alone was sufficient to promote spontaneous fusion. In addition, biochemical differentiation, assessed by expression of placental alkaline phosphatase, was upregulated in caveolin-1 siRNA-transfected cells, with or without dbcAMP treatment. Assessment of Akt phosphorylation showed that caveolin-1 knockdown resulted in a significant reduction in phosphorylation at Thr308. Conclusions/Significance Taken together, these results suggest that caveolin-1 regulates BeWo cell differentiation and fusion, possibly through a mechanism involving modulation of Akt activity.
Collapse
Affiliation(s)
- Gavin P Collett
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom.
| | | | | | | |
Collapse
|
24
|
Heterogeneous pathways of maternal-fetal transmission of human viruses (review). Pathol Oncol Res 2010; 15:451-65. [PMID: 19350418 DOI: 10.1007/s12253-009-9166-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 03/24/2009] [Indexed: 12/18/2022]
Abstract
Several viruses can pass the maternal-fetal barrier, and cause diseases of the fetus or the newborn. Recently, however, it became obvious, that viruses may invade fetal cells and organs through different routes without acute consequences. Spermatozoa, seminal fluid and lymphocytes in the sperm may transfer viruses into the human zygotes. Viruses were shown to be integrated into human chromosomes and transferred into fetal tissues. The regular maternal-fetal transport of maternal cells has also been discovered. This transport might implicate that lymphotropic viruses can be released into the fetal organs following cellular invasion. It has been shown that many viruses may replicate in human trophoblasts and syncytiotrophoblast cells thus passing the barrier of the maternal-fetal interface. The transport of viral immunocomplexes had also been suggested, and the possibility has been put forward that even anti-idiotypes mimicking viral epitopes might be transferred by natural mechanisms into the fetal plasma, in spite of the selective mechanisms of apical to basolateral transcytosis in syncytiotrophoblast and basolateral to apical transcytosis in fetal capillary endothelium. The mechanisms of maternal-fetal transcytosis seem to be different of those observed in differentiated cells and tissue cultures. Membrane fusion and lipid rafts of high cholesterol content are probably the main requirements of fetal transcytosis. The long term presence of viruses in fetal tissues and their interactions with the fetal immune system might result in post partum consequences as far as increased risk of the development of malignancies and chronic pathologic conditions are discussed.
Collapse
|
25
|
Rikihisa Y. Molecular events involved in cellular invasion by Ehrlichia chaffeensis and Anaplasma phagocytophilum. Vet Parasitol 2009; 167:155-66. [PMID: 19836896 DOI: 10.1016/j.vetpar.2009.09.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ehrlichia chaffeensis and Anaplasma phagocytophilum are obligatory intracellular bacteria that preferentially replicate inside leukocytes by utilizing biological compounds and processes of these primary host defensive cells. These bacteria incorporate cholesterol from the host for their survival. Upon interaction with host monocytes and granulocytes, respectively, these bacteria usurp the lipid raft domain containing GPI-anchored protein to induce a series of signaling events that result in internalization of the bacteria. Monocytes and neutrophils usually kill invading microorganisms by fusion of the phagosomes containing the bacteria with granules containing both antimicrobial peptides and lysosomal hydrolytic enzymes and/or through sequestering vital nutrients. However, E. chaffeensis and A. phagocytophilum alter vesicular traffic to create a unique intracellular membrane-bound compartment that allows their replication in seclusion from lysosomal killing. These bacteria are quite sensitive to reactive oxygen species (ROS), so in order to survive in host cells that are primary mediators of ROS-induced killing, they inhibit activation of NADPH oxidase and assembly of this enzyme in their inclusion compartments. Moreover, host phagocyte activation and differentiation, apoptosis, and IFN-gamma signaling pathways are inhibited by these bacteria. Through reductive evolution, lipopolysaccharide and peptidoglycan that activate the innate immune response, have been eliminated from these gram-negative bacteria at the genomic level. Upon interaction with new host cells, bacterial genes encoding the Type IV secretion apparatus and the two-component regulatory system are up-regulated to sense and adapt to the host environment. Thus dynamic signal transduction events concurrently proceed both in the host cells and in the invading E. chaffeensis and A. phagocytophilum bacteria for successful establishment of intracellular infection. Several bacterial surface-exposed proteins and porins are recently identified. Further functional studies on Ehrlichia and Anaplasma effector or ligand molecules and cognate host cell receptors will undoubtedly advance our understanding of the complex interplay between obligatory intracellular pathogens and their hosts. Such data can be applied towards treatment, diagnosis, and control of ehrlichiosis and anaplasmosis.
Collapse
Affiliation(s)
- Yasuko Rikihisa
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
26
|
Kim SY, Wang TK, Singh RD, Wheatley CL, Marks DL, Pagano RE. Proteomic identification of proteins translocated to membrane microdomains upon treatment of fibroblasts with the glycosphingolipid, C8-β-D
-lactosylceramide. Proteomics 2009; 9:4321-8. [DOI: 10.1002/pmic.200900077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
27
|
Joshi B, Strugnell SS, Goetz JG, Kojic LD, Cox ME, Griffith OL, Chan SK, Jones SJ, Leung SP, Masoudi H, Leung S, Wiseman SM, Nabi IR. Phosphorylated caveolin-1 regulates Rho/ROCK-dependent focal adhesion dynamics and tumor cell migration and invasion. Cancer Res 2008; 68:8210-20. [PMID: 18922892 DOI: 10.1158/0008-5472.can-08-0343] [Citation(s) in RCA: 212] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Rho/ROCK signaling and caveolin-1 (Cav1) are implicated in tumor cell migration and metastasis; however, the underlying molecular mechanisms remain poorly defined. Cav1 was found here to be an independent predictor of decreased survival in breast and rectal cancer and significantly associated with the presence of distant metastasis for colon cancer patients. Rho/ROCK signaling promotes tumor cell migration by regulating focal adhesion (FA) dynamics through tyrosine (Y14) phosphorylation of Cav1. Phosphorylated Cav1 is localized to protrusive domains of tumor cells and Cav1 tyrosine phosphorylation is dependent on Src kinase and Rho/ROCK signaling. Increased levels of phosphorylated Cav1 were associated with elevated GTP-RhoA levels in metastatic tumor cells of various tissue origins. Stable expression and knockdown studies of Cav1 in tumor cells showed that phosphorylated Cav1 expression stimulates Rho activation, stabilizes FAK association with FAs, and promotes cell migration and invasion in a ROCK-dependent and Src-dependent manner. Tyrosine-phosphorylated Cav1, therefore, functions as an effector of Rho/ROCK signaling in the regulation of FA turnover and, thereby, tumor cell migration and invasion. These studies define a feedback loop between Rho/ROCK, Src, and phosphorylated Cav1 in tumor cell protrusions, identifying a novel function for Cav1 in tumor metastasis that may contribute to the poor prognosis of some Cav1-expressing tumors.
Collapse
Affiliation(s)
- Bharat Joshi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
PGAM5 tethers a ternary complex containing Keap1 and Nrf2 to mitochondria. Exp Cell Res 2008; 314:1789-803. [PMID: 18387606 DOI: 10.1016/j.yexcr.2008.02.014] [Citation(s) in RCA: 248] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2007] [Revised: 01/21/2008] [Accepted: 02/21/2008] [Indexed: 11/23/2022]
Abstract
Eukaryote cells balance production of reactive oxygen species (ROS) with levels of anti-oxidant enzyme activity to maintain cellular redox homeostasis. Mitochondria are a major source of ROS, while many anti-oxidant genes are regulated by the Nrf2 transcription factor. Keap1, a redox-regulated substrate adaptor for a cullin-based ubiquitin ligase, targets Nrf2 for proteosome-mediated degradation and represses Nrf2-dependent gene expression. We have previously identified a member of the phosphoglycerate mutase family, PGAM5, as a Keap1-binding protein. In this report, we demonstrate that PGAM5 is targeted to the outer membrane of mitochondria by an N-terminal mitochondrial-localization sequence. Furthermore, we provide evidence that PGAM5 forms a ternary complex containing both Keap1 and Nrf2, in which the dimeric Keap1 protein simultaneously binds both PGAM5 and Nrf2 through their conserved E(S/T)GE motifs. Knockdown of either Keap1 or PGAM5 activates Nrf2-dependent gene expression. We suggest that this ternary complex provides a molecular framework for understanding how nuclear anti-oxidant gene expression is regulated in response to changes in mitochondrial function(s).
Collapse
|
29
|
Mellgren RL. Detergent-resistant membrane subfractions containing proteins of plasma membrane, mitochondrial, and internal membrane origins. ACTA ACUST UNITED AC 2007; 70:1029-36. [PMID: 17870178 DOI: 10.1016/j.jbbm.2007.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2007] [Revised: 07/31/2007] [Accepted: 08/08/2007] [Indexed: 10/22/2022]
Abstract
HEK293 cell detergent-resistant membranes (DRMs) isolated by the standard homogenization protocol employing a Teflon pestle homogenizer yielded a prominent opaque band at approximately 16% sucrose upon density gradient ultracentrifugation. In contrast, cell disruption using a ground glass tissue homogenizer generated three distinct DRM populations migrating at approximately 10%, 14%, and 20% sucrose, named DRM subfractions A, B, and C, respectively. Separation of the DRM subfractions by mechanical disruption suggested that they are physically associated within the cellular environment, but can be dissociated by shear forces generated during vigorous homogenization. All three DRM subfractions possessed cholesterol and ganglioside GM1, but differed in protein composition. Subfraction A was enriched in flotillin-1 and contained little caveolin-1. In contrast, subfractions B and C were enriched in caveolin-1. Subfraction C contained several mitochondrial membrane proteins, including mitofilin and porins. Only subfraction B appeared to contain significant amounts of plasma membrane-associated proteins, as revealed by cell surface labeling studies. A similar distribution of DRM subfractions, as assessed by separation of flotillin-1 and caveolin-1 immunoreactivities, was observed in CHO cells, in 3T3-L1 adipocytes, and in HEK293 cells lysed in detergent-free carbonate. Teflon pestle homogenization of HEK293 cells in the presence of the actin-disrupting agent latrunculin B generated DRM subfractions A-C. The microtubule-disrupting agent vinblastine did not facilitate DRM subfraction separation, and DRMs prepared from fibroblasts of vimentin-null mice were present as a single major band on sucrose gradients, unless pre-treated with latrunculin B. These results suggest that the DRM subfractions are interconnected by the actin cytoskeleton, and not by microtubes or vimentin intermediate filaments. The subfractions described may prove useful in studying discrete protein populations associated with detergent-resistant membranes, and their potential interactions in cell signaling.
Collapse
Affiliation(s)
- Ronald L Mellgren
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine, 3000 Arlington Avenue, Toledo, OH 43614-2598, USA.
| |
Collapse
|
30
|
Thomas V, Fikrig E. Anaplasma phagocytophilum specifically induces tyrosine phosphorylation of ROCK1 during infection. Cell Microbiol 2007; 9:1730-7. [PMID: 17346310 DOI: 10.1111/j.1462-5822.2007.00908.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Anaplasma phagocytophilum, an obligate intracellular pathogen that persists within polymorphonuclear leucocytes, is the second most common tick-borne agent in North America. We now show that infection of a promyelocytic cell line and neutrophils with A. phagocytophilum results in pathogen-specific tyrosine phosphorylation of ROCK1. Phosphorylation is associated with PSGL-1 and Syk, because PSGL-1 blocking antibodies and siRNA targeting Syk interfere with ROCK1 phosphorylation in A. phagocytophilum-infected cells. Knockdown of either Syk or ROCK1 also markedly impaired A. phagocytophilum infection. These data demonstrate a role for A. phagocytophilum-mediated ROCK1 phosphorylation in infection, and suggests that inhibiting this pathway may lead to new, non-antibiotic strategies to treat human granulocytic anaplasmosis.
Collapse
Affiliation(s)
- Venetta Thomas
- Section of Rheumatology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | |
Collapse
|