1
|
Bai X, Wu J, Zhang M, Xu Y, Duan L, Yao K, Zhang J, Bo J, Zhao Y, Xu G, Zu H. DHCR24 Knock-Down Induced Tau Hyperphosphorylation at Thr181, Ser199, Thr231, Ser262, Ser396 Epitopes and Inhibition of Autophagy by Overactivation of GSK3β/mTOR Signaling. Front Aging Neurosci 2021; 13:513605. [PMID: 33967735 PMCID: PMC8098657 DOI: 10.3389/fnagi.2021.513605] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/26/2021] [Indexed: 02/01/2023] Open
Abstract
Accumulating evidences supported that knock-down of DHCR24 is linked to the pathological risk factors of AD, suggesting a potential role of DHCR24 in AD pathogenesis. However, the molecular mechanism link between DHCR24 and tauopathy remains unknown. Here, in order to elucidate the relationship between DHCR24 and tauopathy, we will focus on the effect of DHCR24 on the tau hyperphosphorylation at some toxic sites. In present study, we found that DHCR24 knock-down significantly lead to the hyperphosphorylation of tau sites at Thr181, Ser199, Thr231, Ser262, Ser396. Moreover, DHCR24 knock-down also increase the accumulation of p62 protein, simultaneously decreased the ratio of LC3-II/LC3-I and the number of autophagosome compared to the control groups, suggesting the inhibition of autophagy activity. In contrast, DHCR24 knock-in obviously abolished the effect of DHCR24 knock-down on tau hyperphosphrylation and autophagy. In addition, to elucidate the association between DHCR24 and tauopathy, we further showed that the level of plasma membrane cholesterol, lipid raft-anchored protein caveolin-1, and concomitantly total I class PI3-K (p110α), phospho-Akt (Thr308 and Ser473) were significantly decreased, resulting in the disruption of lipid raft/caveola and inhibition of PI3-K/Akt signaling in silencing DHCR24 SH-SY5Y cells compared to control groups. At the same time, DHCR24 knock-down simultaneously decreased the level of phosphorylated GSK3β at Ser9 (inactive form) and increased the level of phosphorylated mTOR at Ser2448 (active form), leading to overactivation of GSK3β and mTOR signaling. On the contrary, DHCR24 knock-in largely increased the level of membrane cholesterol and caveolin-1, suggesting the enhancement of lipid raft/caveola. And synchronously DHCR24 knock-in also abolished the effect of DHCR24 knock-down on the inhibition of PI3-K/Akt signaling as well as the overactivation of GSK3β and mTOR signaling. Collectively, our data strongly supported DHCR24 knock-down lead to tau hyperphosphorylation and the inhibition of autophagy by a lipid raft-dependent PI3-K/Akt-mediated GSK3β and mTOR signaling. Taking together, our results firstly demonstrated that the decrease of plasma membrane cholesterol mediated by DHCR24 deficiency might contribute to the tauopathy in AD and other tauopathies.
Collapse
Affiliation(s)
- Xiaojing Bai
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Junfeng Wu
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Mengqi Zhang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yixuan Xu
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lijie Duan
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Kai Yao
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jianfeng Zhang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jimei Bo
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yongfei Zhao
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Guoxiong Xu
- The Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China
| | - Hengbing Zu
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Zhou Q, Wu H, Liu Y, Zhang N, Liang H, Gu M, Liu H, Wang H. Effects of different doses of propofol on the growth and expression of PCNA, CD34 and pAKT proteins in xenografted tumor of BALB/C mice with liver cancer. Clin Transl Oncol 2020; 22:1741-1749. [PMID: 32052381 DOI: 10.1007/s12094-020-02311-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/27/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To observe the effects of different doses of propofol on the growth of transplanted liver tumor in BALB/C mice and check the expression of PCNA, CD34 and pAKT proteins to clarify the mechanism on molecule level. METHOD Human primary liver cancer cells SMMC-7721 were subcutaneously cultured in BALB/C mice, and the transplanted tumor model of BALB/C mice was constructed. Forty mice successfully modeled were randomly divided into 5 groups (n = 8): the blank control group (group C), low-fat milk group (group I), low-dose (50 mg/kg) propofol group (P1), middle-dose (100 mg/kg) propofol group (P2) and high dose (150 mg/kg) propofol group (P3). Tumor volume changes were observed at 3, 6, 9, 12, 15 and 18 days (T1, T2, T3, T4, T5, T6 and T7) before and after administration of the drug, and tumor growth curves were plotted. After 19 days of administration, all mice were killed for tumor collection, tumor weight was measured, and the tumor inhibition rate of propofol was calculated. The protein expression of cluster of differentiation 34 (CD34) in transplanted tumor was detected by immunohistochemistry, and the protein expression of proliferating cell nuclear antigen (PCNA) and phospho-Akt (pAKT) was detected by immunofluorescence. RESULTS Compared with group C, there was no significant difference in tumor volume in group I. At T2 ~ 7, the tumor volume of group P1, P2 and P3 decreased successively (P < 0.05). There was no significant difference in the inhibitory rate of tumor in group I, and the inhibitory rate of tumor in group P1, P2 and P3 increased successively (P < 0.05). There was no significant difference in PCNA, CD34, and pAKT protein expression in group I, while PCNA, CD34, and pAKT protein content in P1, P2, P3 groups were successively decreased (P < 0.05). CONCLUSION Propofol had a dose-dependent effect on the growth of liver cancer xenografts in mice, inhibiting the expression of PCNA, CD34 and pAKT proteins, and the effect was most obvious in the 150 mg/kg propofol group.
Collapse
Affiliation(s)
- Q Zhou
- Department of Anaesthesiology, Affiliated Foshan Hospital of Sun Yat-sen University, 81# North, Linnandadao Road, Chancheng, Foshan City, 528000, Guangdong Province, China.
| | - H Wu
- Department of Anaesthesiology, Affiliated Foshan Hospital of Sun Yat-sen University, 81# North, Linnandadao Road, Chancheng, Foshan City, 528000, Guangdong Province, China
| | - Y Liu
- Department of Anesthesiology, LinZi People's Hospital, Linzi, Shandong, China
| | - N Zhang
- Department of Anesthesiology, LinZi People's Hospital, Linzi, Shandong, China
| | - H Liang
- Department of Anaesthesiology, Affiliated Foshan Hospital of Sun Yat-sen University, 81# North, Linnandadao Road, Chancheng, Foshan City, 528000, Guangdong Province, China
| | - M Gu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - H Liu
- Department of Anaesthesiology, Affiliated Foshan Hospital of Sun Yat-sen University, 81# North, Linnandadao Road, Chancheng, Foshan City, 528000, Guangdong Province, China
| | - H Wang
- Department of Anaesthesiology, Affiliated Foshan Hospital of Sun Yat-sen University, 81# North, Linnandadao Road, Chancheng, Foshan City, 528000, Guangdong Province, China
| |
Collapse
|
3
|
Zhai L, Liu Y, Zhao W, Chen Q, Guo T, Wei W, Luo Z, Huang Y, Ma C, Huang F, Dai X. Aerobic and resistance training enhances endothelial progenitor cell function via upregulation of caveolin-1 in mice with type 2 diabetes. Stem Cell Res Ther 2020; 11:10. [PMID: 31900223 PMCID: PMC6942272 DOI: 10.1186/s13287-019-1527-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 11/26/2019] [Accepted: 12/10/2019] [Indexed: 12/30/2022] Open
Abstract
Background To explore the effect of aerobic training (AT), resistance training (RT) or a combination of AT and RT (AT+RT) on the function of endothelial progenitor cells (EPCs) in mice with type 2 diabetes and the potential effective mechanisms Methods Eight-week-old db/db male mice were used as type 2 diabetic animal models in this study. Mice were randomly assigned to the control group (n = 5), AT group (n = 5), RT group (n = 5) and AT+RT group (n = 5). Mice in the control group remained sedentary with no specific training requirement. Mice were motivated to perform AT, RT or AT+RT by a gentle pat on their body for 3 or 4 days/week for 14 days. AT was performed by treadmill running, RT was performed by ladder climbing and AT+RT involved both AT and RT. Bone-derived EPCs were isolated after 14 days of the intervention. EPC expression of CD31, CD34, CD133, CD144 and VEGFR2 was detected by immunofluorescence staining. Fluorescence detection was performed on attached mononuclear cells to detect double-positive EPCs. We then explored the effect of caveolin-1 knockdown (lentiviral vector with caveolin-1-siRNA) on the proliferation and adherence of EPCs and the concentration of caveolin-1 and PI3K/AKT via western blot analyses. Results Compared to the mice in the control group, the mice in the AT, RT and AT+RT groups presented significant increases in proliferation and adherence after 14 days of intervention. AT+RT induced an increase in EPC adherence, which was greater than that of the control, RT and AT groups. Caveolin-1 knockdown inhibited the EPC proliferative and adherent abilities. The AT+RT group showed higher levels of caveolin-1 and p-AKT than the control group, but these changes were decreased by caveolin-1-siRNA transfection. Conclusion Combined AT and RT is an effective way to improve EPC function through upregulation of caveolin-1 in mice with type 2 diabetes.
Collapse
Affiliation(s)
- Lu Zhai
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Yuhua Liu
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Wenpiao Zhao
- Department of Nursing, Guangxi JiangBin Hospital, Nanning, 530021, China
| | - Qingyun Chen
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Tao Guo
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Wei Wei
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Zhuchun Luo
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Department of Internal Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Yanfeng Huang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Cui Ma
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Xia Dai
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
4
|
Krndija D, Fairhead M. IGF1R undergoes active and directed centripetal transport on filopodia upon receptor activation. Biochem J 2019; 476:3583-3593. [PMID: 31738383 DOI: 10.1042/bcj20190665] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/06/2019] [Accepted: 11/18/2019] [Indexed: 11/17/2022]
Abstract
Filopodia are thin, actin-based membrane protrusions with roles in sensing external mechanical and chemical cues, such as growth factor gradients in tissues. It was proposed that the chemical sensing role of filopodia is achieved through clearance of activated signaling receptors from filopodia. Type I insulin-like growth factor receptor (IGF1R) is a key regulator of normal development and growth, as well as tumor development and progression. Its biological roles depend on its activation upon IGF1 binding at the cell membrane. IGF1R behavior at the cell membrane and in particular in filopodia, has not been established. We found that IGF1 activation led to a gradual reduction in IGF1R puncta in filopodia, and that this clearance depended on actin, non-muscle myosin II, and IGF1R kinase activity. Using single particle tracking of filopodial IGF1R, we established that ligand-free IGF1R undergoes non-directional unidimensional diffusion along the filopodium. Moreover, after initial diffusion, the ligand-bound IGF1R is actively transported along the filopodium towards the filopodium base, and consequently cleared from the filopodium. Our results show that IGF1R can move directionally on the plasma membrane protrusions, supporting a sensory role for filopodia in interpreting local IGF1 gradients.
Collapse
Affiliation(s)
- Denis Krndija
- Department of Biochemistry, University of Oxford, Oxford, U.K
| | | |
Collapse
|
5
|
Beneficial effects of spermidine on cardiovascular health and longevity suggest a cell type-specific import of polyamines by cardiomyocytes. Biochem Soc Trans 2018; 47:265-272. [PMID: 30578348 DOI: 10.1042/bst20180622] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/16/2018] [Accepted: 11/21/2018] [Indexed: 01/04/2023]
Abstract
Recent and exciting in vivo studies show that supplementation with the polyamine spermidine (Spd) is cardioprotective and prolongs lifespan in both mice and humans. The mechanisms behind Spd-induced cardioprotection are supposed to involve Spd-evoked stimulation of autophagy, mitophagy and mitochondrial respiration and improved the mechano-elastical function of cardiomyocytes. Although cellular uptake of Spd was not characterized, these results suggest that Spd is imported by the cardiomyocytes and acts intracellularly. In the light of these new and thrilling data, we discuss in the present review cellular polyamine import with a special focus on mechanisms that may be relevant for Spd uptake by electrically excitable cells such as cardiomyocytes.
Collapse
|
6
|
Jain N, Kalailingam P, Tan KW, Tan HB, Sng MK, Chan JSK, Tan NS, Thanabalu T. Conditional knockout of N-WASP in mouse fibroblast caused keratinocyte hyper proliferation and enhanced wound closure. Sci Rep 2016; 6:38109. [PMID: 27909303 PMCID: PMC5133560 DOI: 10.1038/srep38109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 11/04/2016] [Indexed: 12/15/2022] Open
Abstract
Neural-Wiskott Aldrich Syndrome Protein (N-WASP) is expressed ubiquitously, regulates actin polymerization and is essential during mouse development. We have previously shown that N-WASP is critical for cell-ECM adhesion in fibroblasts. To characterize the role of N-WASP in fibroblast for skin development, we generated a conditional knockout mouse model in which fibroblast N-WASP was ablated using the Cre recombinase driven by Fibroblast Specific Protein promoter (Fsp-Cre). N-WASPFKO (N-WASPfl/fl; Fsp-cre) were born following Mendelian genetics, survived without any visible abnormalities for more than 1 year and were sexually reproductive, suggesting that expression of N-WASP in fibroblast is not critical for survival under laboratory conditions. Histological sections of N-WASPFKO mice skin (13 weeks old) showed thicker epidermis with higher percentage of cells staining for proliferation marker (PCNA), suggesting that N-WASP deficient fibroblasts promote keratinocyte proliferation. N-WASPFKO mice skin had elevated collagen content, elevated expression of FGF7 (keratinocyte growth factor) and TGFβ signaling proteins. Wound healing was faster in N-WASPFKO mice compared to control mice and N-WASP deficient fibroblasts were found to have enhanced collagen gel contraction properties. These results suggest that N-WASP deficiency in fibroblasts improves wound healing by growth factor-mediated enhancement of keratinocyte proliferation and increased wound contraction in mice.
Collapse
Affiliation(s)
- Neeraj Jain
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Pazhanichamy Kalailingam
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Kai Wei Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Hui Bing Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Ming Keat Sng
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Jeremy Soon Kiat Chan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Nguan Soon Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore.,Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Agency for Science Technology &Research, 138673, Singapore.,KK Research Centre, KK Women's and Children's Hospital, 100 Bukit Timah Road, 229899, Singapore
| | - Thirumaran Thanabalu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| |
Collapse
|
7
|
Perez-Diaz S, Garcia-Rodriguez B, Gonzalez-Irazabal Y, Valero M, Lagos-Lizan J, Arbones-Mainar JM. Knockdown of PTRF ameliorates adipocyte differentiation and functionality of human mesenchymal stem cells. Am J Physiol Cell Physiol 2016; 312:C83-C91. [PMID: 27856429 DOI: 10.1152/ajpcell.00246.2016] [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: 08/22/2016] [Revised: 11/09/2016] [Accepted: 11/09/2016] [Indexed: 01/09/2023]
Abstract
Healthy expansion of human adipose tissue requires mesenchymal stem cells (hMSC) able to proliferate and differentiate into mature adipocytes. Hence, characterization of those factors that coordinate hMSC-to-adipocyte transition is of paramount importance to modulate the adipose tissue expansion. It has been previously reported that the adipogenic program of hMSC can be disrupted by upregulating caveolar proteins, and polymerase I and transcript release factor (PTRF) is an integral component of caveolae, highly expressed in adipose tissue. Here, we hypothesized that the role of PTRF in adipocyte functionality might stem from an effect on hMSC. To test this hypothesis, we isolated hMSC from the subcutaneous fat depot. We found an upregulated expression of the PTRF associated with decreased adipogenic potential of hMSC, likely due to the existence of senescent adipocyte precursors. Employing short hairpin RNA-based constructs to stably reduce PTRF, we were able to restore insulin sensitivity and reduced basal lipolysis and leptin levels in human adipocytes with high levels of PTRF. Additionally, we pinpointed the detrimental effect caused by PTRF on the adipose tissue to the existence of senescent adipocyte precursors unable to proliferate and differentiate into adipocytes. This study provides evidence that impaired adipocyte functionality can be corrected, at least partially, by PTRF downregulation and warrants further in vivo research in patients with dysfunctional adipose tissue to prevent metabolic complications.
Collapse
Affiliation(s)
- Sergio Perez-Diaz
- Adipocyte and Fat Biology Laboratory (AdipoFat), Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Instituto Aragonés de Ciencias de la Salud (IACS), Instituto de Investigación Sanitaria (IIS) Aragón, Zaragoza, Spain
| | | | | | - Monica Valero
- Unidad de Cirugía, Hospital Royo Villanova, Zaragoza, Spain; and
| | | | - Jose M Arbones-Mainar
- Adipocyte and Fat Biology Laboratory (AdipoFat), Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Instituto Aragonés de Ciencias de la Salud (IACS), Instituto de Investigación Sanitaria (IIS) Aragón, Zaragoza, Spain; .,CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Boothe T, Lim GE, Cen H, Skovsø S, Piske M, Li SN, Nabi IR, Gilon P, Johnson JD. Inter-domain tagging implicates caveolin-1 in insulin receptor trafficking and Erk signaling bias in pancreatic beta-cells. Mol Metab 2016; 5:366-378. [PMID: 27110488 PMCID: PMC4837300 DOI: 10.1016/j.molmet.2016.01.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 01/18/2016] [Accepted: 01/25/2016] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE The role and mechanisms of insulin receptor internalization remain incompletely understood. Previous trafficking studies of insulin receptors involved fluorescent protein tagging at their termini, manipulations that may be expected to result in dysfunctional receptors. Our objective was to determine the trafficking route and molecular mechanisms of functional tagged insulin receptors and endogenous insulin receptors in pancreatic beta-cells. METHODS We generated functional insulin receptors tagged with pH-resistant fluorescent proteins between domains. Confocal, TIRF and STED imaging revealed a trafficking pattern of inter-domain tagged insulin receptors and endogenous insulin receptors detected with antibodies. RESULTS Surprisingly, interdomain-tagged and endogenous insulin receptors in beta-cells bypassed classical Rab5a- or Rab7-mediated endocytic routes. Instead, we found that removal of insulin receptors from the plasma membrane involved tyrosine-phosphorylated caveolin-1, prior to trafficking within flotillin-1-positive structures to lysosomes. Multiple methods of inhibiting caveolin-1 significantly reduced Erk activation in vitro or in vivo, while leaving Akt signaling mostly intact. CONCLUSIONS We conclude that phosphorylated caveolin-1 plays a role in insulin receptor internalization towards lysosomes through flotillin-1-positive structures and that caveolin-1 helps bias physiological beta-cell insulin signaling towards Erk activation.
Collapse
Affiliation(s)
- Tobias Boothe
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Gareth E Lim
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Haoning Cen
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Søs Skovsø
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Micah Piske
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Shu Nan Li
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ivan R Nabi
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Patrick Gilon
- Pôle d'endocrinologie, diabète et nutrition, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium
| | - James D Johnson
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
9
|
Joglekar M, Elbezanti WO, Weitzman MD, Lehman HL, van Golen KL. Caveolin-1 mediates inflammatory breast cancer cell invasion via the Akt1 pathway and RhoC GTPase. J Cell Biochem 2016; 116:923-33. [PMID: 25559359 DOI: 10.1002/jcb.25025] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/18/2014] [Indexed: 12/23/2022]
Abstract
With a propensity to invade the dermal lymphatic vessels of the skin overlying the breast and readily metastasize, inflammatory breast cancer (IBC) is arguably the deadliest form of breast cancer. We previously reported that caveolin-1 is overexpressed in IBC and that RhoC GTPase is a metastatic switch responsible for the invasive phenotype. RhoC-driven invasion requires phosphorylation by Akt1. Using a reliable IBC cell line we set out to determine if caveolin-1 expression affects RhoC-mediated IBC invasion. Caveolin-1 was down regulated by introduction of siRNA or a caveolin scaffolding domain. The ability of the cells to invade was tested and the status of Akt1 and RhoC GTPase examined. IBC cell invasion is significantly decreased when caveolin-1 is down regulated. Activation of Akt1 is decreased when caveolin-1 is down regulated, leading to decreased phosphorylation of RhoC GTPase. Thus, we report here that caveolin-1 overexpression mediates IBC cell invasion through activation Akt1, which phosphorylates RhoC GTPase.
Collapse
Affiliation(s)
- Madhura Joglekar
- Department of Biological Sciences, The Center for Translational Cancer Research, The University of Delaware, Newark, Delaware; The Helen F. Graham Cancer Center, Newark, Delaware
| | | | | | | | | |
Collapse
|
10
|
Grossi M, Rippe C, Sathanoori R, Swärd K, Forte A, Erlinge D, Persson L, Hellstrand P, Nilsson BO. Vascular smooth muscle cell proliferation depends on caveolin-1-regulated polyamine uptake. Biosci Rep 2014; 34:e00153. [PMID: 25301005 PMCID: PMC4240025 DOI: 10.1042/bsr20140140] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 10/09/2014] [Indexed: 12/11/2022] Open
Abstract
Much evidence highlights the importance of polyamines for VSMC (vascular smooth muscle cell) proliferation and migration. Cav-1 (caveolin-1) was recently reported to regulate polyamine uptake in intestinal epithelial cells. The aim of the present study was to assess the importance of Cav-1 for VSMC polyamine uptake and its impact on cell proliferation and migration. Cav-1 KO (knockout) mouse aortic cells showed increased polyamine uptake and elevated proliferation and migration compared with WT (wild-type) cells. Both Cav-1 KO and WT cells expressed the smooth muscle differentiation markers SM22 and calponin. Cell-cycle phase distribution analysis revealed a higher proportion of Cav-1 KO than WT cells in the S phase. Cav-1 KO cells were hyper-proliferative in the presence but not in the absence of extracellular polyamines, and, moreover, supplementation with exogenous polyamines promoted proliferation in Cav-1 KO but not in WT cells. Expression of the solute carrier transporters Slc7a1 and Slc43a1 was higher in Cav-1 KO than in WT cells. ODC (ornithine decarboxylase) protein and mRNA expression as well as ODC activity were similar in Cav-1 KO and WT cells showing unaltered synthesis of polyamines in Cav-1 KO cells. Cav-1 was reduced in migrating cells in vitro and in carotid lesions in vivo. Our data show that Cav-1 negatively regulates VSMC polyamine uptake and that the proliferative advantage of Cav-1 KO cells is critically dependent on polyamine uptake. We provide proof-of-principle for targeting Cav-1-regulated polyamine uptake as a strategy to fight unwanted VSMC proliferation as observed in restenosis.
Collapse
Key Words
- caveolin-1
- cell cycle
- ornithine decarboxylase
- polyamine transporter
- polyamine
- vascular smooth muscle cell
- asmc, aortic smooth muscle cell
- cav-1, caveolin-1
- cea, carotid endarterectomy
- dfmo, difluoromethylornithine
- dmem, dulbecco’s modified eagle’s medium
- hbss, hanks balanced salt solution
- [3h]put, [3h]putrescine
- hrp, horseradish peroxidise
- [3h]spd, [3h]spermidine
- hsp90, heat-shock protein 90
- ko, knockout
- odc, ornithine decarboxylase
- pi, propidium iodide
- qrt-pcr, quantitative real-time pcr
- vsmc, vascular smooth muscle cell
- wt, wild-type
Collapse
MESH Headings
- Amino Acid Transport Systems, Basic/genetics
- Amino Acid Transport Systems, Basic/metabolism
- Animals
- Blotting, Western
- Calcium-Binding Proteins/metabolism
- Carotid Arteries/metabolism
- Carotid Arteries/surgery
- Caveolin 1/genetics
- Caveolin 1/metabolism
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- DNA/biosynthesis
- Gene Expression
- Immunohistochemistry
- Mice, Inbred C57BL
- Mice, Knockout
- Microfilament Proteins/metabolism
- Muscle Proteins/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Ornithine Decarboxylase/genetics
- Ornithine Decarboxylase/metabolism
- Polyamines/metabolism
- Polyamines/pharmacokinetics
- Polyamines/pharmacology
- Rats, Wistar
- Reverse Transcriptase Polymerase Chain Reaction
- Calponins
Collapse
Affiliation(s)
- Mario Grossi
- *Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Catarina Rippe
- *Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Ramasri Sathanoori
- †Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Karl Swärd
- *Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Amalia Forte
- ‡Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - David Erlinge
- †Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Lo Persson
- *Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Per Hellstrand
- *Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Bengt-Olof Nilsson
- *Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
11
|
Marissal-Arvy N, Duron E, Parmentier F, Zizzari P, Mormède P, Epelbaum J. QTLs influencing IGF-1 levels in a LOU/CxFischer 344F2 rat population. Tracks towards the metabolic theory of Ageing. Growth Horm IGF Res 2013; 23:220-228. [PMID: 24028904 DOI: 10.1016/j.ghir.2013.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 04/29/2013] [Accepted: 08/12/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Since a reduction of the insulin/IGF-1 signaling cascade extends life span in many species and IGF-1 signaling might partly mediate the effects of caloric restriction (CR), an experimental intervention for increasing longevity, the purpose of the present study was to use quantitative trait loci (QTL) analysis, an unbiased genetic approach, to identify particular regions of the genome influencing plasma IGF-1 levels in an F2 intercross between F344 and LOU/C rats; the latter being an inbred strain of Wistar origin, considered as a model of healthy aging since it resists to age (and diet)-induced obesity. DESIGN F1 hybrids were obtained by crossbreeding LOU/C with F344 rats, and then F1 were bred inter se to obtain the F2 population, of which 93 males and 94 females were studied. Total plasma IGF-1 levels were determined by radioimmunoassay. A genome scan of the F2 population was made with 100 microsatellite markers) selected for their polymorphism between LOU/C and F344 strains (and by covering evenly the whole genome. RESULTS By simple interval mapping sex-dependent QTLs were found on chromosome 17 in males and on chromosome 18 in females. By multiple interval mapping, additional QTLs were found on chromosomes 1, 4, 5, 6, 12, 15 and 19 in males and on chromosomes 3, 5, 6, 12 and 17 in females. Only the markers D1Rat196 and D12Mgh5 were found in both males and females. The majority of QTLs corresponded to metabolic syndrome (cardiac function: n = 45 (30%), obesity/diabetes: n = 22 (15%), inflammation: n = 19 (13%) and only a limited number to body weight: n = 13 (9%), proliferation (n = 10 (7%) or ossification: n = 7 (5%). Ninety-six candidate genes were located on the different QTLs. A significant proportion of these genes are connected to IGF-1 production and receptor pathways (n = 18) or metabolic syndrome (n = 11). CONCLUSIONS Subsequent studies are necessary to determine whether the genetic networks underscored are also involved in age-associated obesity, diabetes and inflammation as well as cardiovascular impairments.
Collapse
Affiliation(s)
- Nathalie Marissal-Arvy
- INRA, Laboratory of Nutrition and Integrative Neurobiology, UMR1286, 33076 Bordeaux Cedex, France; Univ. Bordeaux, Laboratory of Nutrition and Integrative Neurobiology, UMR1286, 33076 Bordeaux Cedex, France
| | | | | | | | | | | |
Collapse
|
12
|
Yang SJ, Chen CY, Chang GD, Wen HC, Chen CY, Chang SC, Liao JF, Chang CH. Activation of Akt by advanced glycation end products (AGEs): involvement of IGF-1 receptor and caveolin-1. PLoS One 2013; 8:e58100. [PMID: 23472139 PMCID: PMC3589465 DOI: 10.1371/journal.pone.0058100] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 02/03/2013] [Indexed: 01/08/2023] Open
Abstract
Diabetes is characterized by chronic hyperglycemia, which in turn facilitates the formation of advanced glycation end products (AGEs). AGEs activate signaling proteins such as Src, Akt and ERK1/2. However, the mechanisms by which AGEs activate these kinases remain unclear. We examined the effect of AGEs on Akt activation in 3T3-L1 preadipocytes. Addition of AGEs to 3T3-L1 cells activated Akt in a dose- and time-dependent manner. The AGEs-stimulated Akt activation was blocked by a PI3-kinase inhibitor LY 294002, Src inhibitor PP2, an antioxidant NAC, superoxide scavenger Tiron, or nicotinamide adenine dinucleotide phosphate (NAD(P)H) oxidase inhibitor DPI, suggesting the involvement of Src and NAD(P)H oxidase in the activation of PI3-kinase-Akt pathway by AGEs. AGEs-stimulated Src tyrosine phosphorylation was inhibited by NAC, suggesting that Src is downstream of NAD(P)H oxidase. The AGEs-stimulated Akt activity was sensitive to Insulin-like growth factor 1 receptor (IGF-1R) kinase inhibitor AG1024. Furthermore, AGEs induced phosphorylation of IGF-1 receptorβsubunit (IGF-1Rβ) on Tyr1135/1136, which was sensitive to PP2, indicating that AGEs stimulate Akt activity by transactivating IGF-1 receptor. In addition, the AGEs-stimulated Akt activation was attenuated by β-methylcyclodextrin that abolishes the structure of caveolae, and by lowering caveolin-1 (Cav-1) levels with siRNAs. Furthermore, addition of AGEs enhanced the interaction of phospho-Cav-1 with IGF-1Rβ and transfection of 3T3-L1 cells with Cav-1 Y14F mutants inhibited the activation of Akt by AGEs. These results suggest that AGEs activate NAD(P)H oxidase and Src which in turn phosphorylates IGF-1 receptor and Cav-1 leading to activation of IGF-1 receptor and the downstream Akt in 3T3-L1 cells. AGEs treatment promoted the differentiation of 3T3-L1 preadipocytes and addition of AG1024, LY 294002 or Akt inhibitor attenuated the promoting effect of AGEs on adipogenesis, suggesting that IGF-1 receptor, PI3-Kinase and Akt are involved in the facilitation of adipogenesis by AGEs.
Collapse
Affiliation(s)
- Su-Jung Yang
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Chen-Yu Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Miaoli, Taiwan, Republic of China
| | - Geen-Dong Chang
- Graduate Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Hui-Chin Wen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Miaoli, Taiwan, Republic of China
| | - Ching-Yu Chen
- Department of Family Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Geriatric Research, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Miaoli, Taiwan, Republic of China
| | - Shi-Chuan Chang
- Chest Department, Taipei Veterans General Hospital, Institute of Emergency and Critical Care Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- * E-mail: (SCC); (CHC)
| | - Jyh-Fei Liao
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Chung-Ho Chang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Miaoli, Taiwan, Republic of China
- Ph.D. Program for Aging, College of Medicine, China Medical University, Taichung, Taiwan, Republic of China
- * E-mail: (SCC); (CHC)
| |
Collapse
|
13
|
Hamoudane M, Maffioli S, Cordera R, Maggi D, Salani B. Caveolin-1 and polymerase I and transcript release factor: new players in insulin-like growth factor-I receptor signaling. J Endocrinol Invest 2013; 36:204-8. [PMID: 23404184 DOI: 10.3275/8848] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Caveolae are plasma membrane regions enriched in Caveolin proteins which regulate vesicular transport, endocytosis, and cell signaling. IGF-I receptor (IGF-IR) localizes in caveolae and tyrosine phosphorylates Caveolin-1 (Cav-1), the most represented caveolar protein. Cav-1 participates to IGF-IR internalization and signaling directly interacting with IGF-IR and its substrates. Recently, polymerase I and transcript release factor (PTRF) or Cavin-1, has been identified in the caveolar backbone. PTRF does not play a Cav-1 ancillary role and emerging data support a direct role of PTRF in IGF-IR signaling. PTRF and Cav-1 can bind IGF-IR and regulate IGF-IR internalization and plasma membrane replacement, mechanisms frequently deregulated in cancer cells. Although the exact roles of Cav-1 and IGF-IR in human cancer continue to be a matter of some debate, there is a strong evidence for an association between Cav-1 and IGF-IR in cancer development. With the discovery of IGF-IR interaction with PTRF in caveolae, new insight emerged to understand the growing functions of these domains in IGF-I action.
Collapse
Affiliation(s)
- M Hamoudane
- Department of Internal Medicine (DiMI) University of Genoa, Genoa, Italy
| | | | | | | | | |
Collapse
|
14
|
Tahir SA, Yang G, Goltsov A, Song KD, Ren C, Wang J, Chang W, Thompson TC. Caveolin-1-LRP6 signaling module stimulates aerobic glycolysis in prostate cancer. Cancer Res 2013; 73:1900-11. [PMID: 23302227 DOI: 10.1158/0008-5472.can-12-3040] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Caveolin 1 (Cav-1) is a plasma membrane-associated protein with the capacity to modulate signaling activities in a context-dependent fashion. Interactions between Cav-1 and low-density lipoprotein receptor-related protein 6 (LRP6) were reported to be important for the regulation of Wnt-β-catenin (β-cat) signaling. Cav-1 also interacts with insulin and IGF-I receptors (IGF-IR/IR) and can stimulate IR kinase activities. We found positive correlation between Cav-1 and LRP6 expression in both human primary prostate cancer and metastasis tissues and in PC-3 cells. Cav-1 stimulation of Wnt-β-cat signaling and c-Myc levels was positively associated with LRP6 expression in LNCaP, PC-3, and DU145 prostate cancer cells. Importantly, LRP6 and, to a lesser extent, Cav-1 were found to stimulate aerobic glycolysis. These activities were positively associated with the expression of HK2 and Glut3 and shown to be dependent on Akt signaling by both gene knockdown and chemical inhibition methods. We further showed that Cav-1 and LRP6 exert their effects on Akt and glycolytic activities by stimulating IGF-IR/IR signaling. Overall, our results show that Cav-1 interacts with LRP6 to generate an integrated signaling module that leads to the activation of IGF-IR/IR and results in stimulation of Akt-mTORC1 signaling and aerobic glycolysis in prostate cancer.
Collapse
Affiliation(s)
- Salahaldin A Tahir
- Department of Genitourinary Medical Oncology-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Filippin-Monteiro FB, de Oliveira EM, Sandri S, Knebel FH, Albuquerque RC, Campa A. Serum amyloid A is a growth factor for 3T3-L1 adipocytes, inhibits differentiation and promotes insulin resistance. Int J Obes (Lond) 2011; 36:1032-9. [PMID: 21986708 PMCID: PMC3419975 DOI: 10.1038/ijo.2011.193] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND/OBJECTIVES Serum amyloid A (SAA) is an acute-phase protein that has been recently correlated with obesity and insulin resistance. Therefore, we first examined whether human recombinant SAA (rSAA) could affect the proliferation, differentiation and metabolism of 3T3-L1 preadipocytes. DESIGN Preadipocytes were treated with rSAA and analyzed for changes in viability and [³H-methyl]-thymidine incorporation as well as cell cycle perturbations using flow cytometry analysis. The mRNA expression profiles of adipogenic factors during the differentiation protocol were also analyzed using real-time PCR. After differentiation, 2-deoxy-[1,2-³H]-glucose uptake and glycerol release were evaluated. RESULTS rSAA treatment caused a 2.6-fold increase in cell proliferation, which was consistent with the results from flow cytometry showing that rSAA treatment augmented the percentage of cells in the S phase (60.9±0.54%) compared with the control cells (39.8±2.2%, (***) P<0.001). The rSAA-induced cell proliferation was mediated by the ERK1/2 signaling pathway, which was assessed by pretreatment with the inhibitor PD98059. However, the exposure of 3T3-L1 cells to rSAA during the differentiation process resulted in attenuated adipogenesis and decreased expression of adipogenesis-related factors. During the first 72 h of differentiation, rSAA inhibited the differentiation process by altering the mRNA expression kinetics of adipogenic transcription factors and proteins, such as PPARγ2 (peroxisome proliferator-activated receptor γ 2), C/EBPβ (CCAAT/enhancer-binding protein β) and GLUT4. rSAA prevented the intracellular accumulation of lipids and, in fully differentiated cells, increased lipolysis and prevented 2-deoxy-[1,2-³H]-glucose uptake, which favors insulin resistance. Additionally, rSAA stimulated the secretion of proinflammatory cytokines interleukin 6 and tumor necrosis factor α, and upregulated SAA3 mRNA expression during adipogenesis. CONCLUSIONS We showed that rSAA enhanced proliferation and inhibited differentiation in 3T3-L1 preadipocytes and altered insulin sensitivity in differentiated cells. These results highlight the complex role of SAA in the adipogenic process and support a direct link between obesity and its co-morbidities such as type II diabetes.
Collapse
|
16
|
Mo S, Yang S, Cui Z. New glimpses of caveolin-1 functions in embryonic development and human diseases. FRONTIERS IN BIOLOGY 2011; 6:367. [PMID: 32215005 PMCID: PMC7089126 DOI: 10.1007/s11515-011-1132-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 12/30/2010] [Indexed: 11/17/2022]
Abstract
Caveolin-1 (Cav-1) isoforms, including Cav-1α and Cav-1β, were identified as integral membrane proteins and the major components of caveolae. Cav-1 proteins are highly conserved during evolution from {itCaenorhabditis elegans} to human and are capable of interacting with many signaling molecules through their caveolin scaffolding domains to regulate the activities of multiple signaling pathways. Thus, Cav-1 plays crucial roles in the regulation of cellular proliferation, differentiation and apoptosis in a cell-specific and contextual manner. In addition, Cav-1 is essential for embryonic development of vertebrates owing to its regulation of BMP, Wnt, TGF-β and other key signaling molecules. Moreover, Cav-1 is mainly expressed in terminally differentiated cells and its abnormal expression is often associated with human diseases, such as tumor progression, cardiovascular diseases, fibrosis, lung regeneration, and diseases related to virus. In this review, we will further discuss the potential of Cav-1 as a target for disease therapy and multiple drug resistance.
Collapse
Affiliation(s)
- Saijun Mo
- Department of Basic Oncology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001 China
| | - Shengli Yang
- Department of Basic Oncology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001 China
| | - Zongbin Cui
- Key Laboratory of Biodiversity and Conservation of Aquatic Organism, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 China
| |
Collapse
|
17
|
Park JH, Ryu JM, Han HJ. Involvement of caveolin-1 in fibronectin-induced mouse embryonic stem cell proliferation: role of FAK, RhoA, PI3K/Akt, and ERK 1/2 pathways. J Cell Physiol 2010; 226:267-75. [PMID: 20658539 DOI: 10.1002/jcp.22338] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Fibronectin (FN) is the foremost proliferation-associated extracellular matrix component promoting cell adhesion, migration, and survival. We examined the effect of FN on cell proliferation and the related signaling pathways in mouse embryonic stem (ES) cells. FN increased integrin β1, Src, focal adhesion kinase (FAK), and caveolin-1 phosphorylation levels in a time-dependent manner. Phosphorylation of Src, FAK, and caveolin-1 was attenuated by integrin β1 neutralizing antibody. Integrin β1, Src, and FAK coimmunoprecipitated with caveolin-1 in the presence of FN. In addition, FN increased RhoA and Rho kinase activation, which were completely blocked by PP2, FAK small interfering RNA (siRNA), caveolin-1 siRNA, or the caveolar disruptor methyl-β-cyclodextrin (MβCD). FN also increased phosphorylation of Akt and ERK 1/2, which were significantly blocked by either FAK siRNA, caveolin-1 siRNA, MβCD, GGTI-286 (RhoA inhibitor), or Y-27632 (Rho kinase inhibitor). FN-induced increase of protooncogenes (c-fos, c-myc, and c-Jun) and cell-cycle regulatory proteins (cyclin D1/CDK4 and cyclin E/CDK2) expression levels were attenuated by FAK siRNA or caveolin-1 siRNA. Furthermore, inhibition of each pathway such as integrin β1, Src, FAK, caveolin-1, RhoA, Akt, and ERK 1/2 blocked FN-induced [(3)H]-thymidine incorporation. We conclude that FN stimulates mouse ES cell proliferation via RhoA-PI3K/Akt-ERK 1/2 pathway through caveolin-1 phosphorylation.
Collapse
Affiliation(s)
- Jae Hong Park
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK 21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | | | | |
Collapse
|
18
|
Cleveland-Donovan K, Maile LA, Tsiaras WG, Tchkonia T, Kirkland JL, Boney CM. IGF-I activation of the AKT pathway is impaired in visceral but not subcutaneous preadipocytes from obese subjects. Endocrinology 2010; 151:3752-63. [PMID: 20555032 PMCID: PMC2940538 DOI: 10.1210/en.2010-0043] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Obesity morbidity is associated with excess visceral adiposity, whereas sc adipose tissue is much less metabolically hazardous. Human abdominal sc preadipocytes have greater capacity for proliferation, differentiation, and survival than omental preadipocytes. IGF-I is a critical mediator of preadipocyte proliferation, differentiation, and survival through multiple signaling pathways. We investigated IGF-I action in primary cultures of human preadipocytes isolated from sc and omental adipose tissue of obese subjects. IGF-I-stimulated DNA synthesis was significantly lower in omental compared with sc preadipocytes. IGF-I phosphorylation of the IGF-I receptor and the ERK pathway was comparable in sc and omental cells. However, omental preadipocytes had decreased insulin receptor substrate (IRS)-1 protein associated with increased IRS-1-serine(636/639) phosphorylation and degradation. IGF-I-stimulated phosphorylation of AKT on serine(473) but not threonine(308) was decreased in omental cells, and activation of downstream targets, including S6Kinase, glycogen synthase kinase-3, and Forkhead box O1 was also impaired. CyclinD1 abundance was decreased in omental cells due to increased degradation. Over-expression of IRS-1 by lentivirus in omental preadipocytes increased IGF-I-stimulated AKT-serine(473) phosphorylation. The mammalian target of rapamycin (mTOR)-Rictor complex regulates phosphorylation of AKT-serine(473) in 3T3-L1 adipocytes, but knockdown of Rictor by lentivirus-delivered short hairpin RNA in sc preadipocytes did not affect AKT-serine(473) phosphorylation by IGF-I. These data reveal an intrinsic defect in IGF-I activation of the AKT pathway in omental preadipocytes from obese subjects that involves IRS-1 but probably not mTOR-Rictor complex. We conclude that impaired cell cycle regulation by AKT contributes to the distinct growth phenotype of preadipocytes in visceral fat of obese subjects.
Collapse
Affiliation(s)
- Kelly Cleveland-Donovan
- Department of Pediatrics, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | | | | | | | | | | |
Collapse
|
19
|
D'Souza A, Fordjour L, Ahmad A, Cai C, Kumar D, Valencia G, Aranda JV, Beharry KD. Effects of probiotics, prebiotics, and synbiotics on messenger RNA expression of caveolin-1, NOS, and genes regulating oxidative stress in the terminal ileum of formula-fed neonatal rats. Pediatr Res 2010; 67:526-31. [PMID: 20101198 DOI: 10.1203/pdr.0b013e3181d4ff2b] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Necrotizing enterocolitis (NEC) afflicts extremely low birth weight neonates, and probiotics reduces its incidence and severity. NO is involved in the pathogenesis of NEC, and caveolin-1 regulates NO signaling. We tested the hypothesis that intestinal caveolin-1 and NOS are deficient in formula-fed neonatal rats and that supplementation with "Florastar Kids" and/or galacto-oligosaccharides and fructo-oligosaccharides preserves caveolin-1 and NOS. At birth (P0), neonatal rat pups were maternally fed or hand-gavaged with or without supplemented formula. Samples from the terminal ileum were analyzed for total NO metabolites, growth factors, and gene expression of caveolin-1, NOS isoforms, and antioxidants. Our data showed that formula feeding with and without supplementation resulted in significant growth restriction. Despite suboptimal nutrition, growth factors involved in intestinal repair and regeneration were increased in the neonatal rat ileum. Caveolin-1, endothelial NOS, and neuronal NOS were simultaneously down-regulated with formula feeding while inducible NOS was up-regulated. Superoxide dismutase and glutathione peroxidase were up-regulated with supplementation. These data provide a probable mechanism for the benefits of supplemented formula for decreasing the severity of NEC by preserving the antioxidant systems.
Collapse
Affiliation(s)
- Antoni D'Souza
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, New York 11203, USA. ad'
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Wu H, Jiang H, Lu D, Xiong Y, Qu C, Zhou D, Mahmood A, Chopp M. Effect of simvastatin on glioma cell proliferation, migration, and apoptosis. Neurosurgery 2010; 65:1087-96; discussion 1096-7. [PMID: 19934968 DOI: 10.1227/01.neu.0000360130.52812.1d] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE In this study, we investigated the effects of simvastatin on proliferation, migration, and apoptosis in human U251 and U87 glioma cells and the underlying molecular mechanism. METHODS We used colony formation assay to test the cell proliferation, in vitro scratch assay to examine the cell migration, and caspase-3 activity assay, annexin V staining, and cytochrome C release to evaluate the cell apoptosis. Lipid raft fractions were isolated from glioma cells. Total cholesterol content assay was used to test the change of cholesterol level in lipid raft fractions. Immunocytochemistry staining was performed to detect the changes of lipid rafts in cell membranes. Western blotting analysis was performed to examine the signal transduction both in cells and in lipid raft fractions. RESULTS Simvastatin inhibited proliferation and migration of U251 and U87 cells dose dependently. Simvastatin induced an increase of caspase-3 activity and annexin V staining, and down-regulated the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Simvastatin also decreased cholesterol content in lipid raft fractions, suppressed caveolin-1 expression in the lipid rafts, and induced Fas translocation into lipid rafts, suggesting that simvastatin may inhibit the prosurvival PI3K/Akt pathway and trigger caspase-3-dependent apoptotic cell death through the modulation of lipid rafts. CONCLUSION These results suggest that modulation of lipid rafts, Fas translocation, and PI3K/Akt/caspase-3 pathway are involved in the antitumor effect of simvastatin and may have a potential role in cancer prevention and treatment.
Collapse
Affiliation(s)
- Hongtao Wu
- Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan 48202, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Hacquemand R, Buron G, Pourié G, Karrer M, Jacquot L, Brand G. Effects of CO2 inhalation exposure on mice vomeronasal epithelium. Cell Biol Toxicol 2009; 26:309-17. [PMID: 19924548 DOI: 10.1007/s10565-009-9143-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 11/03/2009] [Indexed: 10/20/2022]
Abstract
Nasal epitheliums are the first sites of the respiratory tract in contact with the external environment and may therefore be susceptible to damage from exposure to many toxic volatile substances (i.e., volatile organic components, vapors, and gases). In the field of inhalation toxicology, a number of studies have considered the main olfactory epithelium, but few have dealt with the epithelium of the vomeronasal organ (VNO). However, in several species such as in rodents, the VNO (an organ of pheromone detection) plays an important role in social interactions, and alterations of this organ are known to induce adaptative behavioral disturbances. Among volatile toxicants, health effects of inhaled gases have been thoroughly investigated, especially during CO(2) inhalation because of its increasing atmospheric concentration. Therefore, this work was designed to examine the effects of 3% CO(2) inhalation on VNO in two different exposure conditions (5 h/day and 12 h/day) in mice. Behavioral sensitivity tests to urine of congener and histological measurements of VNO were conducted before, during (weeks 1-4), and after (weeks 5-8) CO(2) inhalation exposures. Results showed no significant modifications of behavioral responses to urine, but there were significant changes of both cell number and thickness of the VNO epithelium. Moreover, the findings indicated a selectively dose-dependent effect of CO(2), and further research could use other gases in the same manner for comparison.
Collapse
Affiliation(s)
- Romain Hacquemand
- Laboratoire de Neurosciences, Université de Franche-Comté, Besançon, France
| | | | | | | | | | | |
Collapse
|
22
|
Shen X, Xi G, Radhakrishnan Y, Clemmons DR. Identification of novel SHPS-1-associated proteins and their roles in regulation of insulin-like growth factor-dependent responses in vascular smooth muscle cells. Mol Cell Proteomics 2009; 8:1539-51. [PMID: 19299420 DOI: 10.1074/mcp.m800543-mcp200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tyrosine phosphatase non-receptor type substrate-1 (SHPS-1), a transmembrane protein, plays a vital role in cell migration and proliferation. Our previous studies have shown that insulin-like growth factor-I (IGF-I) stimulates SHPS-1 phosphorylation, leading to recruitment of SHP-2, c-Src, Shc, and Grb2.p85 to phosphorylated SHPS-1. Assembly of this signaling complex is required for optimal stimulation of both mitogen-activated protein and phosphatidylinositol 3-kinase pathways. The main aim of the present study was to identify novel proteins that interacted with the cytoplasmic domain of SHPS-1 (SHPS-1/CD) in response to IGF-I stimulation and define the role of these interactions in mediating specific biological functions. We performed a functional proteomic screening to identify SHPS-1 binding partners using combination of mRNA display and the tandem affinity purification-tag methods. Screening identified a number of proteins not previously known to interact with phosphorylated SHPS-1/CD. These novel SHPS-1 binding partners represent several functional categories including heat shock proteins, protein kinases and phosphatases, and proteins that regulate transcription or translation. In Vivo and in vitro studies suggested that most of the proteins bound to SHPS-1 via binding to one of the four SH2 domain containing proteins, SHP-2, CTK, SUPT6H, and STAT1, that directly bound to SHPS-1. Although the binding of most of these proteins to SHPS-1 was positively regulated by IGF-I, a few were negatively regulated, suggesting differential regulation of protein complexes assembled on SHPS-1/CD in response to IGF-I. Further studies showed that truncation of SHPS-1/CD significantly impaired IGF-I-dependent AKT signal transduction and subsequent biological functions including cell survival, protein synthesis, protein aggregation, and prevention of apoptosis. The results emphasize the importance of formation of SHPS-1 signaling complex induced by IGF-I and provide novel insights into our knowledge of the role of this molecular scaffold in regulation of IGF-I-stimulated signal transduction and biological actions.
Collapse
Affiliation(s)
- Xinchun Shen
- Department of Medicine, University of North Carolina, School of Medicine, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
23
|
Romanenko VG, Roser KS, Melvin JE, Begenisich T. The role of cell cholesterol and the cytoskeleton in the interaction between IK1 and maxi-K channels. Am J Physiol Cell Physiol 2009; 296:C878-88. [PMID: 19176762 DOI: 10.1152/ajpcell.00438.2008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Recently, we demonstrated a novel interaction between large-conductance (maxi-K or K(Ca)1.1) and intermediate-conductance (IK1 or K(Ca)3.1) Ca(2+)-activated K channels: activation of IK1 channels causes the inhibition of maxi-K activity (Thompson J and Begenisich T. J Gen Physiol 127: 159-169, 2006). Here we show that the interaction between these two channels can be regulated by the membrane cholesterol level in parotid acinar cells. Depletion of cholesterol using methyl-beta-cyclodextrin weakened, while cholesterol enrichment increased, the ability of IK1 activation to inhibit maxi-K channels. Cholesterol's stereoisomer, epicholesterol, was unable to substitute for cholesterol in the interaction between the two K channels, suggesting a specific cholesterol-protein interaction. This suggestion was strengthened by the results of experiments in which cholesterol was replaced by coprostanol and epicoprostanol. These two sterols have nearly identical effects on membrane physical properties and cholesterol-rich microdomain stability, but had very different effects on the IK1/maxi-K interaction. In addition, the IK1/maxi-K interaction was unaltered in cells lacking caveolin, the protein essential for formation and stability of caveolae. Finally, disruption of the actin cytoskeleton restored the IK1-induced maxi-K inhibition that was lost with cell cholesterol depletion, demonstrating the importance of an intact cytoskeleton for the cholesterol-dependent regulation of the IK1/maxi-K interaction.
Collapse
Affiliation(s)
- Victor G Romanenko
- Dept. of Pharmacology, Box 711, Univ. of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|