1
|
Gould M, Potapov D, Nicholson H. Location and Movement of the Oxytocin Receptor Differ Between the Normal and Diseased Prostate. Cancers (Basel) 2025; 17:182. [PMID: 39857963 PMCID: PMC11763401 DOI: 10.3390/cancers17020182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/09/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND In normal prostate cells, receptors for oxytocin (OT), a peptide involved in regulating prostate growth are sequestered within membrane microdomains called caveolae. During cancer progression, polymerase-transcript-release factor (PTRF) is downregulated, caveolae structures are lost and receptors move onto the cell membrane. This study investigated whether proteins responsible for caveolae formation were affected by the OT peptide, also, how OT treatment affected oxytocin receptor (OTR) movement within living cells. METHODS Normal human prostate epithelial cells (PrEC) expressing caveolin and PTRF and androgen-independent (PC3) cancer cells expressing caveolin but not PTRF were used. OTR, caveolin and PTRF expression was determined in human prostate tissue. RESULTS PTRF expression decreased in tissue alongside an increase in malignancy. Caveolin-1 expression was downregulated by OT treatment. Caveolin-2 was decreased by OT in PrEC cells but increased in PC3 cells. PTRF was decreased by OT in PrEC. TIRF microscopy showed OTR translocated from caveolae to caveolae in normal cells, whereas OTR moved without restraint in malignant cells, possibly stimulating signaling pathways. CONCLUSIONS This study provides evidence for the ability of OT to regulate caveolin and PTRF expression. This study elucidates possible mechanisms by which cell receptors and caveolae proteins interact to enhance cell proliferation.
Collapse
Affiliation(s)
- Maree Gould
- Department of Anatomy, University of Otago, P.O. Box 913, Dunedin 9054, New Zealand;
| | - Daniil Potapov
- Department of Physiology, University of Otago, P.O. Box 913, Dunedin 9054, New Zealand;
| | - Helen Nicholson
- Department of Anatomy, University of Otago, P.O. Box 913, Dunedin 9054, New Zealand;
| |
Collapse
|
2
|
Bustamante A, Baritaki S, Zaravinos A, Bonavida B. Relationship of Signaling Pathways between RKIP Expression and the Inhibition of EMT-Inducing Transcription Factors SNAIL1/2, TWIST1/2 and ZEB1/2. Cancers (Basel) 2024; 16:3180. [PMID: 39335152 PMCID: PMC11430682 DOI: 10.3390/cancers16183180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Untreated primary carcinomas often lead to progression, invasion and metastasis, a process that involves the epithelial-to-mesenchymal transition (EMT). Several transcription factors (TFs) mediate the development of EMT, including SNAIL1/SNAIL2, TWIST1/TWIST2 and ZEB1/ZEB2, which are overexpressed in various carcinomas along with the under expression of the metastasis suppressor Raf Kinase Inhibitor Protein (RKIP). Overexpression of RKIP inhibits EMT and the above associated TFs. We, therefore, hypothesized that there are inhibitory cross-talk signaling pathways between RKIP and these TFs. Accordingly, we analyzed the various properties and biomarkers associated with the epithelial and mesenchymal tissues and the various molecular signaling pathways that trigger the EMT phenotype such as the TGF-β, the RTK and the Wnt pathways. We also presented the various functions and the transcriptional, post-transcriptional and epigenetic regulations for the expression of each of the EMT TFs. Likewise, we describe the transcriptional, post-transcriptional and epigenetic regulations of RKIP expression. Various signaling pathways mediated by RKIP, including the Raf/MEK/ERK pathway, inhibit the TFs associated with EMT and the stabilization of epithelial E-Cadherin expression. The inverse relationship between RKIP and the TF expressions and the cross-talks were further analyzed by bioinformatic analysis. High mRNA levels of RKIP correlated negatively with those of SNAIL1, SNAIL2, TWIST1, TWIST2, ZEB1, and ZEB2 in several but not all carcinomas. However, in these carcinomas, high levels of RKIP were associated with good prognosis, whereas high levels of the above transcription factors were associated with poor prognosis. Based on the inverse relationship between RKIP and EMT TFs, it is postulated that the expression level of RKIP in various carcinomas is clinically relevant as both a prognostic and diagnostic biomarker. In addition, targeting RKIP induction by agonists, gene therapy and immunotherapy will result not only in the inhibition of EMT and metastases in carcinomas, but also in the inhibition of tumor growth and reversal of resistance to various therapeutic strategies. However, such targeting strategies must be better investigated as a result of tumor heterogeneities and inherent resistance and should be better adapted as personalized medicine.
Collapse
Affiliation(s)
- Andrew Bustamante
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Stavroula Baritaki
- Laboratory of Experimental Oncology, Division of Surgery, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Apostolos Zaravinos
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus
| | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
An Z, Tian J, Zhao X, Liu L, Yang X, Zhang M, Zhang L, Song X. Regulation of cardiovascular and cardiac functions by caveolins. FEBS J 2024; 291:3753-3761. [PMID: 37060249 DOI: 10.1111/febs.16798] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/16/2023]
Abstract
Caveolae are intracellular vesicles with diameters ranging from 50 to 100 nm. The role of caveolins in mediating oxidative stress, autophagy, apoptosis, fibrosis, and vascular remodeling has attracted increasing attention in cardiovascular therapy. Several studies have suggested that caveolin could be a therapeutic target for the treatment of cardiac and/or vascular injury via several pathophysiological mechanisms. Despite substantial advances in our understanding of the basic biology of vesicles over the past decade, the relevance and specific role of these mechanisms in cardiovascular homeostasis remains ambiguous. Here, we review the macroscopic role of caveolins in protecting cardiac function and, at the microscopic level, examine possible cardioprotective caveolar mechanisms, including their antioxidative stress, antiapoptosis, autophagy-regulatory, antifibrosis, and angiogenesis-promoting properties. We believe that the role of caveolins in cardiac functioning has not been fully elucidated and is an important line of future research with several cardioprotective implications.
Collapse
Affiliation(s)
- Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Xueyao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mingduo Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lijun Zhang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Patritti-Cram J, Rahrmann EP, Rizvi TA, Scheffer KC, Phoenix TN, Largaespada DA, Ratner N. NF1-dependent disruption of the blood-nerve-barrier is improved by blockade of P2RY14. iScience 2024; 27:110294. [PMID: 39100928 PMCID: PMC11294707 DOI: 10.1016/j.isci.2024.110294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/17/2023] [Accepted: 06/14/2024] [Indexed: 08/06/2024] Open
Abstract
The blood-nerve-barrier (BNB) that regulates peripheral nerve homeostasis is formed by endoneurial capillaries and perineurial cells surrounding the Schwann cell (SC)-rich endoneurium. Barrier dysfunction is common in human tumorigenesis, including in some nerve tumors. We identify barrier disruption in human NF1 deficient neurofibromas, which were characterized by reduced perineurial cell glucose transporter 1 (GLUT1) expression and increased endoneurial fibrin(ogen) deposition. Conditional Nf1 loss in murine SCs recapitulated these alterations and revealed decreased tight junctions and decreased caveolin-1 (Cav1) expression in mutant nerves and in tumors, implicating reduced Cav1-mediated transcytosis in barrier disruption and tumorigenesis. Additionally, elevated receptor tyrosine kinase activity and genetic deletion of Cav1 increased endoneurial fibrin(ogen), and promoted SC tumor formation. Finally, when SC lacked Nf1, genetic loss or pharmacological inhibition of P2RY14 rescued Cav1 expression and barrier function. Thus, loss of Nf1 in SC causes dysfunction of the BNB via P2RY14-mediated G-protein coupled receptor (GPCR) signaling.
Collapse
Affiliation(s)
- Jennifer Patritti-Cram
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0713, USA
| | - Eric P. Rahrmann
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Tilat A. Rizvi
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Katherine C. Scheffer
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Timothy N. Phoenix
- Division of Pharmaceutical Sciences, James L. Wrinkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
| | - David A. Largaespada
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
5
|
Jiang X, Wu S, Hu C. A narrative review of the role of exosomes and caveolin-1 in liver diseases and cancer. Int Immunopharmacol 2023; 120:110284. [PMID: 37196562 DOI: 10.1016/j.intimp.2023.110284] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/16/2023] [Accepted: 05/02/2023] [Indexed: 05/19/2023]
Abstract
Exosomes are nanoscale (40-100 nm) vesicles secreted by different types of cells and have attracted extensive interest in recent years because of their unique role in disease development. It can carry related goods, such as lipids, proteins, and nucleic acids, to mediate intercellular communication. This review summarizes exosome biogenesis, release, uptake, and their role in mediating the development of liver diseases and cancer, such as viral hepatitis, drug-induced liver injury, alcohol-related liver disease, non-alcoholic fatty liver disease, hepatocellular carcinoma, and other tumors. Meanwhile, a fossa structural protein, caveolin-1(CAV-1), has also been proposed to be involved in the development of various diseases, especially liver diseases and tumors. In this review, we discuss the role of CAV-1 in liver diseases and different tumor stages (inhibition of early growth and promotion of late metastasis) and the underlying mechanisms by which CAV-1 regulates the process. In addition, CAV-1 has also been found to be a secreted protein that can be released directly through the exosome pathway or change the cargo composition of the exosomes, thus contributing to enhancing the metastasis and invasion of cancer cells during the late stage of tumor development. In conclusion, the role of CAV-1 and exosomes in disease development and the association between them remains to be one challenging uncharted area.
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
| | - 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
| | - 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
|
Cigremis Y, Ozen H, Durhan M, Tunc S, Kose E. Effects of caffeic acid phenethyl ester use and inhibition of p42/44 MAP kinase signal pathway on caveolin 1 gene expression and antioxidant system in chronic renal failure model of rats. Drug Chem Toxicol 2023; 46:197-208. [PMID: 34957889 DOI: 10.1080/01480545.2021.2016043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Effects of Caffeic acid phenethyl ester (CAPE) and/or PD98059 (PD) on the gene expression of Caveolin-1 (CAV1) and reduced glutathione (GSH), malondialdehyde (MDA), copper-zinc superoxide dismutase (CuZn-SOD), and catalase (CAT) enzyme activities were investigated in an experimental chronic renal failure model in rats. Eighty Wistar rats were divided into eight groups for a 28-day study: Control, CsA (Cyclosporine A), CsA-V (CsA solvent), CsA + PD (CsA + PD98059), CsA + PD + CAPE, CsA + CAPE, CAPE-V (CAPE solvent), and PD-V (PD98059 solvent). Serum blood urea nitrogen and creatinine levels, as well as histopathological findings indicated the development of renal failure in the CsA group. Kidney GSH levels decreased while MDA levels, CuZn-SOD, and CAT activities increased significantly in the CsA group compared to control indicating oxidative stress. CAV1 gene expression significantly decreased in the CsA group compared to the control. PD98059 and CAPE applications made positive improvements in the levels of the parameters investigated. PD98059 and CAPE applications in CsA given animals increased GSH and CAV1 gene expressions and decreased CuZn-SOD and CAT levels compared to the CsA group. In conclusion, it was shown that PD98059 and CAPE could attenuate the effects of chronic renal failure, and CAV1 is suggested as a therapeutic target and the inhibition of the p44/42 MAPK pathway may be a new approach for the treatment of renal degenerations.
Collapse
Affiliation(s)
- Yilmaz Cigremis
- Department of Medical Biology and Genetics, Medical Faculty, Inonu University, Malatya, Turkey
| | - Hasan Ozen
- Department of Pathology, Faculty of Veterinary Medicine, Balıkesir University, Balıkesir, Turkey
| | - Merve Durhan
- Department of Medical Biology and Genetics, Medical Faculty, Inonu University, Malatya, Turkey
| | - Selahattin Tunc
- Department of Medical Biology and Genetics, Medical Faculty, Inonu University, Malatya, Turkey
| | - Evren Kose
- Department of Anatomy, Medical Faculty, Inonu University, Malatya, Turkey
| |
Collapse
|
7
|
Abou-Fadel J, Bhalli M, Grajeda B, Zhang J. CmP Signaling Network Leads to Identification of Prognostic Biomarkers for Triple-Negative Breast Cancer in Caucasian Women. Genet Test Mol Biomarkers 2022; 26:198-219. [PMID: 35481969 DOI: 10.1089/gtmb.2021.0221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective: Triple-negative breast cancer (TNBC) constitutes ∼15% of all diagnosed invasive breast cancer cases with limited options for treatment since immunotherapies that target ER, PR, and HER2 receptors are ineffective. Progesterone (PRG) can induce its effects through either classic, nonclassic, or combined responses by binding to classic nuclear PRG receptors (nPRs) or nonclassic membrane PRG receptors (mPRs). Under PRG-induced actions, we previously demonstrated that the CCM signaling complex (CSC) can couple both nPRs and mPRs into a CmPn signaling network, which plays an important role during nPR(+) breast cancer tumorigenesis. We recently defined the novel CmP signaling network in African American women (AAW)-derived TNBC cells, which overlapped with our previously defined CmPn network in nPR(+) breast cancer cells. Methods: Under mPR-specific steroid actions, we measured alterations to key tumorigenic pathways in Caucasian American women (CAW)- derived TNBC cells, with RNAseq/proteomic and systems biology approaches. Exemption from ethics approval from IRB: This study only utilized cultured NBC cell lines with publicly available TNBC clinical data sets. Results: Our results demonstrated that TNBCs in CAW share similar altered signaling pathways, as TNBCs in AAW, under mPR-specific steroid actions, demonstrating the overall aggressive nature of TNBCs, regardless of racial differences. Furthermore, in this report, we have deconvoluted the CmP signalosome, using systems biology approaches and CAW-TNBC clinical data, to identify 21 new CAW-TNBC-specific prognostic biomarkers that reinforce the definitive role of CSC and mPR signaling during CAW-TNBC tumorigenesis. Conclusion: This new set of potential prognostic biomarkers may revolutionize molecular mechanisms and currently known concepts of tumorigenesis in CAW-TNBCs, leading to hopeful new therapeutic strategies.
Collapse
Affiliation(s)
- Johnathan Abou-Fadel
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, Texas, USA
| | - Muaz Bhalli
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, Texas, USA
| | - Brian Grajeda
- Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas, USA
| | - Jun Zhang
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, Texas, USA
| |
Collapse
|
8
|
Goyal A, Agrawal N, Jain A, Gupta JK, Garabadu D. Role of caveolin-eNOS platform and mitochondrial ATP-sensitive potassium channel in abrogated cardioprotective effect of ischemic preconditioning in postmenopausal women. BRAZ J PHARM SCI 2022; 58. [DOI: 10.1590/s2175-97902022e20081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
| | | | - Ankit Jain
- Dr. Hari Singh Gour Central University, India
| | | | | |
Collapse
|
9
|
Russell JS, Griffith TA, Peart JN, Headrick JP. Cardiomyoblast caveolin expression: effects of simulated diabetes, α-linolenic acid, and cell signaling pathways. Am J Physiol Cell Physiol 2020; 319:C11-C20. [PMID: 32348174 DOI: 10.1152/ajpcell.00499.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Caveolins regulate myocardial substrate handling, survival signaling, and stress resistance; however, control of expression is incompletely defined. We test how metabolic features of type 2 diabetes (T2D), and modulation of cell signaling, influence caveolins in H9c2 cardiomyoblasts. Cells were exposed to glucose (25 vs. 5 mM), insulin (100 nM), or palmitate (0.1 mM), individually or combined, and the effects of adenylate cyclase (AC) activation (50 μM forskolin), focal adhesion kinase (FAK) or protein kinase C β2 (PKCβ2) inhibition (1 μM FAK inhibitor 14 or CGP-53353, respectively) or the polyunsaturated fatty acid (PUFA) α-linolenic acid (ALA; 10 μM) were tested. Simulated T2D (elevated glucose + insulin + palmitate) depressed caveolin-1 and -3 without modifying caveolin-2. Caveolin-3 repression was primarily palmitate dependent, whereas high glucose (HG) and insulin independently increased caveolin-3 (while reducing expression when combined). Differential control was evident: baseline caveolin-3 was suppressed by FAK/PKCβ2 and insensitive to AC activities, with baseline caveolin-1 and -2 suppressed by AC and insensitive to FAK/PKCβ2. Forskolin and ALA selectively preserved caveolin-3 in T2D cells, whereas PKCβ2 and FAK inhibition increased caveolin-3 under all conditions. Despite preservation of caveolin-3, ALA did not modify nucleosome content (apoptosis marker) or transcription of proinflammatory mediators in T2D cells. In summary, caveolin-1 and -3 are strongly repressed with simulated T2D, with caveolin-3 particularly sensitive to palmitate; intrinsic PKCβ2 and FAK activities depress caveolin-3 in healthy and stressed cells; ALA and AC activation and PKCβ2 inhibition preserve caveolin-3 under T2D conditions; and caveolin-3 changes with T2D and ALA appear unrelated to inflammatory signaling or extent of apoptosis.
Collapse
Affiliation(s)
- Jake S Russell
- School of Medical Science, Griffith University Gold Coast, Southport, Queensland, Australia
| | - Tia A Griffith
- School of Medical Science, Griffith University Gold Coast, Southport, Queensland, Australia
| | - Jason N Peart
- School of Medical Science, Griffith University Gold Coast, Southport, Queensland, Australia
| | - John P Headrick
- School of Medical Science, Griffith University Gold Coast, Southport, Queensland, Australia
| |
Collapse
|
10
|
Kreisler AD, Terranova MJ, Somkuwar SS, Purohit DC, Wang S, Head BP, Mandyam CD. In vivo reduction of striatal D1R by RNA interference alters expression of D1R signaling-related proteins and enhances methamphetamine addiction in male rats. Brain Struct Funct 2020; 225:1073-1088. [PMID: 32246242 DOI: 10.1007/s00429-020-02059-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/18/2020] [Indexed: 01/06/2023]
Abstract
This study sought to determine if reducing dopamine D1 receptor (D1R) expression in the dorsal striatum (DS) via RNA-interference alters methamphetamine self-administration. A lentiviral construct containing a short hairpin RNA (shRNA) was used to knock down D1R expression (D1RshRNA). D1RshRNA in male rats increased responding for methamphetamine (i.v.) under a fixed-ratio schedule in an extended access paradigm, compared to D1R-intact rats. D1RshRNA also produced a vertical shift in a dose-response paradigm and enhanced responding for methamphetamine in a progressive-ratio schedule, generating a drug-vulnerable phenotype. D1RshRNA did not alter responding for sucrose (oral) under a fixed-ratio schedule compared to D1R-intact rats. Western blotting confirmed reduced D1R expression in methamphetamine and sucrose D1RshRNA rats. D1RshRNA reduced the expression of PSD-95 and MAPK-1 and increased the expression of dopamine transporter (DAT) in the DS from methamphetamine, but not sucrose rats. Sucrose density gradient fractionation was performed in behavior-naïve controls, D1RshRNA- and D1R-intact rats to determine the subcellular localization of D1Rs, DAT and D1R signaling proteins. D1Rs, DAT, MAPK-1 and PSD-95 predominantly localized to heavy fractions, and the membrane/lipid raft protein caveolin-1 (Cav-1) and flotillin-1 were distributed equally between buoyant and heavy fractions in controls. Methamphetamine increased localization of PSD-95, Cav-1, and flotillin-1 in D1RshRNA and D1R-intact rats to buoyant fractions. Our studies indicate that reduced D1R expression in the DS increases vulnerability to methamphetamine addiction-like behavior, and this is accompanied by striatal alterations in the expression of DAT and D1R signaling proteins and is independent of the subcellular localization of these proteins.
Collapse
Affiliation(s)
| | | | | | | | - Shanshan Wang
- VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA, 92161, USA
| | - Brian P Head
- VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA, 92161, USA
| | - Chitra D Mandyam
- VA San Diego Healthcare System, San Diego, CA, 92161, USA.
- Department of Anesthesiology, University of California San Diego, San Diego, CA, 92161, USA.
| |
Collapse
|
11
|
Parlakpinar H, Ozhan O, Ermis N, Vardi N, Cigremis Y, Tanriverdi LH, Colak C, Acet A. Acute and Subacute Effects of Low Versus High Doses of Standardized Panax ginseng Extract on the Heart: An Experimental Study. Cardiovasc Toxicol 2019; 19:306-320. [DOI: 10.1007/s12012-019-09512-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
12
|
Santos AL, Preta G. Lipids in the cell: organisation regulates function. Cell Mol Life Sci 2018; 75:1909-1927. [PMID: 29427074 PMCID: PMC11105414 DOI: 10.1007/s00018-018-2765-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/04/2018] [Accepted: 01/29/2018] [Indexed: 12/19/2022]
Abstract
Lipids are fundamental building blocks of all cells and play important roles in the pathogenesis of different diseases, including inflammation, autoimmune disease, cancer, and neurodegeneration. The lipid composition of different organelles can vary substantially from cell to cell, but increasing evidence demonstrates that lipids become organised specifically in each compartment, and this organisation is essential for regulating cell function. For example, lipid microdomains in the plasma membrane, known as lipid rafts, are platforms for concentrating protein receptors and can influence intra-cellular signalling. Lipid organisation is tightly regulated and can be observed across different model organisms, including bacteria, yeast, Drosophila, and Caenorhabditis elegans, suggesting that lipid organisation is evolutionarily conserved. In this review, we summarise the importance and function of specific lipid domains in main cellular organelles and discuss recent advances that investigate how these specific and highly regulated structures contribute to diverse biological processes.
Collapse
Affiliation(s)
- Ana L Santos
- Institut National de la Santé et de la Recherche Médicale, U1001 and Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Giulio Preta
- Institute of Biochemistry, Vilnius University, Sauletekio 7, LT-10257, Vilnius, Lithuania.
| |
Collapse
|
13
|
Díaz-Valdivia NI, Calderón CC, Díaz JE, Lobos-González L, Sepulveda H, Ortíz RJ, Martinez S, Silva V, Maldonado HJ, Silva P, Wehinger S, Burzio VA, Torres VA, Montecino M, Leyton L, Quest AFG. Anti-neoplastic drugs increase caveolin-1-dependent migration, invasion and metastasis of cancer cells. Oncotarget 2017; 8:111943-111965. [PMID: 29340103 PMCID: PMC5762371 DOI: 10.18632/oncotarget.22955] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 11/16/2017] [Indexed: 12/20/2022] Open
Abstract
Expression of the scaffolding protein Caveolin-1 (CAV1) enhances migration and invasion of metastatic cancer cells. Yet, CAV1 also functions as a tumor suppressor in early stages of cancer, where expression is suppressed by epigenetic mechanisms. Thus, we sought to identify stimuli/mechanisms that revert epigenetic CAV1 silencing in cancer cells and evaluate how this affects their metastatic potential. We reasoned that restricted tissue availability of anti-neoplastic drugs during chemotherapy might expose cancer cells to sub-therapeutic concentrations, which activate signaling pathways and the expression of CAV1 to favor the acquisition of more aggressive traits. Here, we used in vitro [2D, invasion] and in vivo (metastasis) assays, as well as genetic and biochemical approaches to address this question. Colon and breast cancer cells were identified where CAV1 levels were low due to epigenetic suppression and could be reverted by treatment with the methyltransferase inhibitor 5’-azacytidine. Exposure of these cells to anti-neoplastic drugs for short periods of time (24-48 h) increased CAV1 expression through ROS production and MEK/ERK activation. In colon cancer cells, increased CAV1 expression enhanced migration and invasion in vitro via pathways requiring Src-family kinases, as well as Rac-1 activity. Finally, elevated CAV1 expression in colon cancer cells following exposure in vitro to sub-cytotoxic drug concentrations increased their metastatic potential in vivo. Therefore exposure of cancer cells to anti-neoplastic drugs at non-lethal drug concentrations induces signaling events and changes in transcription that favor CAV1-dependent migration, invasion and metastasis. Importantly, this may occur in the absence of selection for drug-resistance.
Collapse
Affiliation(s)
- Natalia I Díaz-Valdivia
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Claudia C Calderón
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Jorge E Díaz
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Lorena Lobos-González
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Fundación Ciencia & Vida, Santiago, Chile
| | - Hugo Sepulveda
- Gene Regulation Laboratory, Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile
| | - Rina J Ortíz
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Universidad Bernardo O Higgins, Facultad de Salud, Departamento de Ciencias Químicas y Biológicas, Santiago, Chile
| | - Samuel Martinez
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | | | - Horacio J Maldonado
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Patricio Silva
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Sergio Wehinger
- Faculty of Health Sciences, University of Talca, Interdisciplinary Excellence Research Program Healthy Ageing (PIEI-ES), Talca, Chile
| | - Verónica A Burzio
- Fundación Ciencia & Vida, Santiago, Chile.,Faculty of Biological Sciences, Universidad Andrés Bello, Santiago, Chile
| | - Vicente A Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Martín Montecino
- Gene Regulation Laboratory, Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
14
|
Abstract
The liver is an essential organ for nutrient and drug metabolism - possessing the remarkable ability to sense environmental and metabolic stimuli and provide an optimally adaptive response. Early growth response 1 (Egr1), an immediate early transcriptional factor which acts as a coordinator of the complex response to stress, is induced during liver injury and controls the expression of a wide range of genes involved in metabolism, cell proliferation, and role of Egr1 in liver injury and repair, deficiency of Egr1 delays liver regeneration process. The known upstream regulators of Egr1 include, but are not limited to, growth factors (e.g. transforming growth factor β1, platelet-derived growth factor, epidermal growth factor, hepatocyte growth factor), nuclear receptors (e.g. hepatocyte nuclear factor 4α, small heterodimer partner, peroxisome proliferator-activated receptor-γ), and other transcription factors (e.g. Sp1, E2F transcription factor 1). Research efforts using various animal models such as fatty liver, liver injury, and liver fibrosis contribute greatly to the elucidation of Egr1 function in the liver. Hepatocellular carcinoma (HCC) represents the second leading cause of cancer mortality worldwide due to the heterogeneity and the late stage at which cancer is generally diagnosed. Recent studies highlight the involvement of Egr1 in HCC development. The purpose of this review is to summarize current studies pertaining to the role of Egr1 in liver metabolism and liver diseases including liver cancer.
Collapse
Affiliation(s)
- Nancy Magee
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
15
|
Borsoi C, Leonard F, Lee Y, Zaid M, Elganainy D, Alexander JF, Kai M, Liu YT, Kang Y, Liu X, Koay EJ, Ferrari M, Godin B, Yokoi K. Gemcitabine enhances the transport of nanovector-albumin-bound paclitaxel in gemcitabine-resistant pancreatic ductal adenocarcinoma. Cancer Lett 2017; 403:296-304. [PMID: 28687352 DOI: 10.1016/j.canlet.2017.06.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/03/2017] [Accepted: 06/16/2017] [Indexed: 02/06/2023]
Abstract
The mechanism for improved therapeutic efficacy of the combination therapy with nanoparticle albumin-bound paclitaxel (nAb-PTX) and gemcitabine (gem) for pancreatic ductal adenocarcinoma (PDAC) has been ascribed to enhanced gem transport by nAb-PTX. Here, we used an orthotopic mouse model of gem-resistant human PDAC in which increasing gem transport would not improve the efficacy, thus revealing the importance of nAb-PTX transport. We aimed to evaluate therapeutic outcomes and transport of nAb-PTX to PDAC as a result of (1) encapsulating nAb-PTX in multistage nanovectors (MSV); (2) effect of gem on caveolin-1 expression. Treatment with MSV/nAb-PTX + gem was highly efficient in prolonging animal survival in comparison to other therapeutic regimens. MSV/nAb-PTX + gem also caused a substantial increase in tumor PTX accumulation, significantly reduced tumor growth and tumor cell proliferation, and increased apoptosis. Moreover, gem enhanced caveolin-1 expression in vitro and in vivo, thereby improving transport of nAb-PTX to PDAC. This data was confirmed by analysis of PDACs from patients who received gem-based neo-adjuvant chemotherapy. In conclusion, we found that nAb-PTX treatment of gem-resistant PDAC can be enhanced by (1) gem through up-regulation of caveolin-1 and (2) MSV through increasing accumulation of nAb-PTX in the tumor.
Collapse
Affiliation(s)
- Carlotta Borsoi
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Fransisca Leonard
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Yeonju Lee
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Mohamed Zaid
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Dalia Elganainy
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | | | - Megumi Kai
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Yan Ting Liu
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Yaan Kang
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Eugene J Koay
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA.
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA.
| | - Kenji Yokoi
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA.
| |
Collapse
|
16
|
Methamphetamine reduces expression of caveolin-1 in the dorsal striatum: Implication for dysregulation of neuronal function. Neuroscience 2016; 328:147-56. [PMID: 27138644 DOI: 10.1016/j.neuroscience.2016.04.039] [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: 02/20/2016] [Revised: 03/30/2016] [Accepted: 04/25/2016] [Indexed: 12/15/2022]
Abstract
Role of striatal dopamine D1 receptors (D1Rs) in methamphetamine (Meth) taking and seeking is recognized from contingent Meth self-administration studies. For example, Meth increases levels of D1Rs in the dorsal striatum in animal models of Meth addiction, and blockade of striatal D1Rs decreased responding for Meth and reduced Meth priming-induced drug seeking. However, the mechanism underlying enhanced expression of striatal D1Rs in animals self-administering Meth is unknown and is hypothesized to involve maladaptive intracellular signal transduction mechanism via hyperphosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). D1Rs are predominantly localized to detergent-resistant membrane/lipid raft fractions (MLR fraction), and in vitro studies indicate that D1R signaling and recycling is regulated by the MLR-resident protein caveolin-1 (Cav-1), in an endocytotic-dependent manner. Notably, expression of Cav-1 is inversely regulated by ERK1/2 activation, suggesting a signaling interplay among D1Rs, ERK1/2 and Cav-1. We therefore evaluated the effects of extended access Meth self-administration on expression of striatal D1Rs, activated ERK1/2 and Cav-1. We first report that Cav-1 is heavily expressed in neurons located in the dorsal striatum. We also report that extended access Meth produces compulsive-like unregulated intake of the drug, and these behavioral outcomes are associated with enhanced expression of D1Rs, increased activity of ERK1/2, and reduced Cav-1 expression in the dorsal striatum. These data suggest a possible cellular mechanism that involves Cav-1 regulation of D1R expression in response to escalated Meth intake, and how this response of altered D1Rs and enhanced ERK1/2 activation to Meth self-administration contributes to contingent-related processes such as addiction.
Collapse
|
17
|
Qin L, Yang YB, Yang YX, Zhu N, Liu Z, Ni YG, Li SX, Zheng XL, Liao DF. Inhibition of macrophage-derived foam cell formation by ezetimibe via the caveolin-1/MAPK pathway. Clin Exp Pharmacol Physiol 2016; 43:182-92. [DOI: 10.1111/1440-1681.12524] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 10/13/2015] [Accepted: 12/08/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Li Qin
- Division of Stem Cell Regulation and Application; School of Pharmacy; Hunan University of Chinese Medicine; Changsha Hunan China
| | - Yun-Bo Yang
- Division of Stem Cell Regulation and Application; School of Pharmacy; Hunan University of Chinese Medicine; Changsha Hunan China
- Matthew Mailing Centre for Translational Transplantation Studies; London Health Sciences Centre; Western University; London Canada
| | - Yi-Xin Yang
- Matthew Mailing Centre for Translational Transplantation Studies; London Health Sciences Centre; Western University; London Canada
| | - Neng Zhu
- The First Hospital of Hunan University of Chinese Medicine; Changsha Hunan China
| | - Zheng Liu
- Division of Stem Cell Regulation and Application; School of Pharmacy; Hunan University of Chinese Medicine; Changsha Hunan China
| | - Ya-Guang Ni
- Division of Stem Cell Regulation and Application; School of Pharmacy; Hunan University of Chinese Medicine; Changsha Hunan China
| | - Shun-Xiang Li
- Division of Stem Cell Regulation and Application; School of Pharmacy; Hunan University of Chinese Medicine; Changsha Hunan China
| | - Xi-Long Zheng
- Division of Stem Cell Regulation and Application; School of Pharmacy; Hunan University of Chinese Medicine; Changsha Hunan China
- Department of Biochemistry & Molecular Biology; Libin Cardiovascular Institute of Alberta; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Duan- Fang Liao
- Division of Stem Cell Regulation and Application; School of Pharmacy; Hunan University of Chinese Medicine; Changsha Hunan China
| |
Collapse
|
18
|
Reconciling paradigms of abnormal pulmonary blood flow and quasi-malignant cellular alterations in pulmonary arterial hypertension. Vascul Pharmacol 2016; 83:17-25. [PMID: 26804008 DOI: 10.1016/j.vph.2016.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 01/19/2016] [Indexed: 01/29/2023]
Abstract
In pulmonary arterial hypertension (PAH) structural and functional abnormalities of the small lung vessels interact and lead to a progressive increase in pulmonary vascular resistance and right heart failure. A current pathobiological concept characterizes PAH as a 'quasi-malignant' disease focusing on cancer-like alterations in endothelial cells (EC) and the importance of their acquired apoptosis-resistant, hyper-proliferative phenotype in the process of vascular remodeling. While changes in pulmonary blood flow (PBF) have been long-since recognized and linked to the development of PAH, little is known about a possible relationship between an altered PBF and the quasi-malignant cell phenotype in the pulmonary vascular wall. This review summarizes recognized and hypothetical effects of an abnormal PBF on the pulmonary vascular bed and links these to quasi-malignant changes found in the pulmonary endothelium. Here we describe that abnormal PBF does not only trigger a pulmonary vascular cell growth program, but may also maintain the cancer-like phenotype of the endothelium. Consequently, normalization of PBF and EC response to abnormal PBF may represent a treatment strategy in patients with established PAH.
Collapse
|
19
|
Kitowska A, Wesserling M, Seroczynska B, Szutowicz A, Ronowska A, Peksa R, Pawelczyk T. Differentiation of high-risk stage I and II colon tumors based on evaluation of CAV1 gene expression. J Surg Oncol 2015; 112:408-14. [PMID: 26251082 DOI: 10.1002/jso.23995] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 07/10/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Several molecular markers are currently being investigated for their prognostic or predictive value in colorectal cancer. One of the genes proposed, as a potential molecular marker in CRC is CAV1. METHODS The level of CAV1 expression was investigated in low-stage (I and II TNM) colon cancers using Real-Time PCR and immunohistochemistry. RESULTS The level of CAV1 expression increased in tumors characterized by greater depths of invasiveness. The CAV1 expression level detected in tumors with a depth of invasion at stage T4 was significantly higher compared to that in T2 (P = 0.01) and T3 (P = 0.003) lesions. The length of a patient's survival depended on CAV1 expression level; the 10-year survival rate for patients with elevated expression of CAV1 was ∼59% compared with 91% for patients with reduced or unchanged expression of CAV1 (P = 0.007). The overall survival rate of patients with T3 + T4 lesions was significantly lower (P = 0.006) for patients with tumor displaying elevated CAV1 expression compared with patients with reduced or unchanged CAV1 expression. CONCLUSIONS Evaluation of CAV1 expression offers valuable prognostic information for patients with colorectal cancer, and could be used to select patients with stage I or II disease, who are at increased risk of unfavorable outcomes.
Collapse
Affiliation(s)
- Agnieszka Kitowska
- Department of Molecular Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Martyna Wesserling
- Department of Molecular Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Barbara Seroczynska
- Department of Molecular Medicine, Medical University of Gdansk, Gdansk, Poland.,Central Bank of Tissue and Genetic Material, Medical University of Gdansk, Gdansk, Poland
| | - Andrzej Szutowicz
- Departemnt of Laboratory Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Anna Ronowska
- Departemnt of Laboratory Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Rafal Peksa
- Department of Pathomorphology, Medical University of Gdansk, Gdansk, Poland
| | - Tadeusz Pawelczyk
- Department of Molecular Medicine, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
20
|
Diaz-Valdivia N, Bravo D, Huerta H, Henriquez S, Gabler F, Vega M, Romero C, Calderon C, Owen GI, Leyton L, Quest AFG. Enhanced caveolin-1 expression increases migration, anchorage-independent growth and invasion of endometrial adenocarcinoma cells. BMC Cancer 2015; 15:463. [PMID: 26054531 PMCID: PMC4460862 DOI: 10.1186/s12885-015-1477-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 05/28/2015] [Indexed: 12/28/2022] Open
Abstract
Background Caveolin-1 (CAV1) has been implicated both in tumor suppression and progression, whereby the specific role appears to be context dependent. Endometrial cancer is one of the most common malignancies of the female genital tract; however, little is known about the role of CAV1 in this disease. Methods Here, we first determined by immunohistochemistry CAV1 protein levels in normal proliferative human endometrium and endometrial tumor samples. Then using two endometrial cancer cell lines (ECC: Ishikawa and Hec-1A) we evaluated mRNA and protein levels of CAV1 by real time qPCR and Western blot analysis, respectively. The role of CAV1 expression in ECC malignancy was further studied by either inducing its expression in endometrial cancer cells with the tumor promotor 12-O-tetradecanoyl-phorbol-13-acetate (4β-TPA) or decreasing expression using short-hairpin RNA constructs, and then evaluating the effects of these changes on ECC proliferation, transmigration, matrigel invasion, and colony formation in soft agar. Results Immunohistochemical analysis of endometrial epithelia revealed that substantially higher levels of CAV1 were present in endometrial tumors than the normal proliferative epithelium. Also, in Ishikawa and Hec-1A endometrial cancer cells CAV1 expression was readily detectable. Upon treatment with 4β-TPA CAV1 levels increased and coincided with augmented cell transmigration, matrigel invasion, as well as colony formation in soft agar. Reduction of CAV1 expression using short-hairpin RNA constructs ablated these effects in both cell types whether treated or not with 4β-TPA. Alternatively, CAV1 expression appeared not to modulate significantly proliferation of these cells. Conclusion Our study shows that elevated CAV1, observed in patients with endometrial cancer, is linked to enhanced malignancy of endometrial cancer cells, as evidenced by increased migration, invasion and anchorage-independent growth. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1477-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Natalia Diaz-Valdivia
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile. .,Center for Molecular studies of the Cell (CEMC), Programa de Biologia Celular y Molecular, Instituto de Ciencias Biomedicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Denisse Bravo
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile. .,Facultad de Odontología, Universidad de Chile, Santiago, Chile.
| | - Hernán Huerta
- Center for Molecular studies of the Cell (CEMC), Programa de Biologia Celular y Molecular, Instituto de Ciencias Biomedicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Soledad Henriquez
- Departamento de Obstetricia y Ginecologia, Facultad de Medicina, Hospital Clínico de la Universidad de Chile, Santiago, Chile.
| | - Fernando Gabler
- Departamento de Obstetricia y Ginecologia, Facultad de Medicina, Hospital Clínico de la Universidad de Chile, Santiago, Chile.
| | - Margarita Vega
- Departamento de Obstetricia y Ginecologia, Facultad de Medicina, Hospital Clínico de la Universidad de Chile, Santiago, Chile.
| | - Carmen Romero
- Departamento de Obstetricia y Ginecologia, Facultad de Medicina, Hospital Clínico de la Universidad de Chile, Santiago, Chile.
| | - Claudia Calderon
- Center for Molecular studies of the Cell (CEMC), Programa de Biologia Celular y Molecular, Instituto de Ciencias Biomedicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Gareth I Owen
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Lisette Leyton
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile. .,Center for Molecular studies of the Cell (CEMC), Programa de Biologia Celular y Molecular, Instituto de Ciencias Biomedicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile. .,Center for Molecular studies of the Cell (CEMC), Programa de Biologia Celular y Molecular, Instituto de Ciencias Biomedicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile. .,Laboratorio de Comunicaciones Celulares, Instituto de Ciencias Biomedicas (ICBM). Facultad de Medicina, Universidad de Chile, Av. Indepedencia 1027. Independencia, Santiago, Chile.
| |
Collapse
|
21
|
Abstract
It has been over 20 years since the discovery that caveolar lipid rafts function as signalling organelles. Lipid rafts create plasma membrane heterogeneity, and caveolae are the most extensively studied subset of lipid rafts. A newly emerging paradigm is that changes in caveolae also generate tumour metabolic heterogeneity. Altered caveolae create a catabolic tumour microenvironment, which supports oxidative mitochondrial metabolism in cancer cells and which contributes to dismal survival rates for cancer patients. In this Review, we discuss the role of caveolae in tumour progression, with a special emphasis on their metabolic and cell signalling effects, and their capacity to transform the tumour microenvironment.
Collapse
Affiliation(s)
- Ubaldo E Martinez-Outschoorn
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | - Federica Sotgia
- 1] Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK. [2] Manchester Centre for Cellular Metabolism (MCCM), University of Manchester, Manchester M20 4BX, UK
| | - Michael P Lisanti
- 1] Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK. [2] Manchester Centre for Cellular Metabolism (MCCM), University of Manchester, Manchester M20 4BX, UK
| |
Collapse
|
22
|
Sanders YY, Cui Z, Le Saux CJ, Horowitz JC, Rangarajan S, Kurundkar A, Antony VB, Thannickal VJ. SMAD-independent down-regulation of caveolin-1 by TGF-β: effects on proliferation and survival of myofibroblasts. PLoS One 2015; 10:e0116995. [PMID: 25658089 PMCID: PMC4319960 DOI: 10.1371/journal.pone.0116995] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/17/2014] [Indexed: 12/30/2022] Open
Abstract
Transforming growth factor-β (TGF-β) mediates growth-inhibitory effects on most target cells via activation of the canonical SMAD signaling pathway. This growth-inhibitory activity may be coupled with cellular differentiation. Our studies demonstrate that TGF-β1 inhibits proliferation of primary, non-transformed human lung fibroblasts in association with the induction of myofibroblast differentiation. Differentiated myofibroblasts maintain the capacity to proliferate in response to exogenous mitogenic stimuli and are resistant to serum deprivation-induced apoptosis. These proliferative and anti-apoptotic properties of myofibroblasts are related, in part, to the down-regulation of caveolin-1 (Cav-1) by TGF-β1. Cav-1 down-regulation is mediated by early activation of p38 MAPK and does not require SMAD signaling. In contrast, myofibroblast differentiation is dependent on activation of the SMAD pathway, but not on p38 MAPK. Thus, combinatorial signaling by TGF-β1 of myofibroblast differentiation and down-regulation of Cav-1 by SMAD and p38 MAPK pathways, respectively, confer proliferative and apoptosis-resistant properties to myofibroblasts. Selective targeting of this SMAD-independent, p38-MAPK/Cav-1-dependent pathway is likely to be effective in the treatment of pathological conditions characterized by TGF-β signaling and myofibroblast activation.
Collapse
Affiliation(s)
- Yan Y. Sanders
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Zongbin Cui
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, 48109, United States of America
| | - Claude Jourdan Le Saux
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, United States of America
| | - Jeffrey C. Horowitz
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, 48109, United States of America
| | - Sunad Rangarajan
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Ashish Kurundkar
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Veena B. Antony
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Victor J. Thannickal
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
- * E-mail:
| |
Collapse
|
23
|
Involvement of nitric oxide and caveolins in the age-associated functional and structural changes in a heart under osmotic stress. Biomed Pharmacother 2014; 69:380-7. [PMID: 25661386 DOI: 10.1016/j.biopha.2014.12.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 12/11/2014] [Indexed: 11/22/2022] Open
Abstract
Previous work done in our laboratory showed that water restriction during 24 and 72h induced changes in cardiovascular NOS activity without altering NOS protein levels in young and adult animals. These findings indicate that the involvement of NO in the regulatory mechanisms during dehydration depends on the magnitude of the water restriction and on age. Our aim was to study whether a controlled water restriction of 1 month affects cardiac function, NO synthase (NOS) activity and NOS, and cav-1 and -3 protein levels in rats during aging. Male Sprague-Dawley rats aged 2 and 16 months were divided into 2 groups: (CR) control restriction (WR) water restriction. Measurements of arterial blood pressure, heart rate, oxidative stress, NOS activity and NOS/cav-1 and -3 protein levels were performed. Cardiac function was evaluated by echocardiography. The results showed that adult rats have greater ESV, EDV and SV than young rats with similar SBP. Decreased atria NOS activity was caused by a reduction in NOS protein levels. Adult animals showed increased cav-1. Water restriction decreased NOS activity in young and adult rats associated to an increased cav-1. TBARS levels increased in adult animals. Higher ventricular NOS activity in adulthood would be caused by a reduction in both cav. Water restriction reduced NOS activity and increased cav in both age groups. In conclusion, our results indicated that dehydration modifies cardiac NO system activity and its regulatory proteins cav in order to maintain physiological cardiac function. Functional alterations are induced by the aging process as well as hypovolemic state.
Collapse
|
24
|
Caveolin-1 is required for kinase suppressor of Ras 1 (KSR1)-mediated extracellular signal-regulated kinase 1/2 activation, H-RasV12-induced senescence, and transformation. Mol Cell Biol 2014; 34:3461-72. [PMID: 25002533 DOI: 10.1128/mcb.01633-13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The molecular scaffold kinase suppressor of Ras 1 (KSR1) regulates the activation of the Raf/MEK/extracellular signal-regulated kinase (ERK) signal transduction pathway. KSR1 disruption in mouse embryo fibroblasts (MEFs) abrogates growth factor-induced ERK activation, H-Ras(V12)-induced replicative senescence, and H-Ras(V12)-induced transformation. Caveolin-1 has been primarily described as a major component of the coating structure of caveolae, which can serve as a lipid binding adaptor protein and coordinates the assembly of Ras, Raf, MEK, and ERK. In this study, we show that KSR1 interacts with caveolin-1 and is responsible for MEK and ERK redistribution to caveolin-1-rich fractions. The interaction between KSR1 and caveolin-1 is essential for optimal activation of ERK as a KSR1 mutant unable to interact with caveolin-1 does not efficiently mediate growth factor-induced ERK activation at the early stages of pathway activation. Furthermore, abolishing the KSR1-caveolin-1 interaction increases growth factor demands to promote H-Ras(V12)-induced proliferation and has adverse effects on H-Ras(V12)-induced cellular senescence and transformation. These data show that caveolin-1 is necessary for optimal KSR1-dependent ERK activation by growth factors and oncogenic Ras.
Collapse
|
25
|
Thompson MA, Prakash YS, Pabelick CM. The role of caveolae in the pathophysiology of lung diseases. Expert Rev Respir Med 2013; 8:111-22. [PMID: 24308657 DOI: 10.1586/17476348.2014.855610] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Caveolae are flask-shaped plasma membrane invaginations formed by constitutive caveolin proteins and regulatory cavin proteins. Caveolae harbor a range of signaling components such as receptors, ion channels and regulatory molecules. There is now increasing evidence that caveolins and cavins play an important role in a variety of diseases. However, the mechanisms by which these caveolar proteins affect lung health and disease are still under investigation, with emerging data suggesting complex roles in disease pathophysiology. This review summarizes the current state of understanding of how caveolar proteins contribute to lung structure and function and how their altered expression and/or function can influence lung diseases.
Collapse
|
26
|
Mathew R. Pathogenesis of pulmonary hypertension: a case for caveolin-1 and cell membrane integrity. Am J Physiol Heart Circ Physiol 2013; 306:H15-25. [PMID: 24163076 DOI: 10.1152/ajpheart.00266.2013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pulmonary hypertension (PH) is a progressive disease with a high morbidity and mortality rate. Despite important advances in the field, the precise mechanisms leading to PH are not yet understood. Main features of PH are loss of vasodilatory response, the activation of proliferative and antiapoptotic pathways leading to pulmonary vascular remodeling and obstruction, elevated pressure and right ventricular hypertrophy, resulting in right ventricular failure and death. Experimental studies suggest that endothelial dysfunction may be the key underlying feature in PH. Caveolin-1, a major protein constituent of caveolae, interacts with several signaling molecules including the ones implicated in PH and modulates them. Disruption and progressive loss of endothelial caveolin-1 with reciprocal activation of proliferative pathways occur before the onset of PH, and the rescue of caveolin-1 inhibits proliferative pathways and attenuates PH. Extensive endothelial damage/loss occurs during the progression of the disease with subsequent enhanced expression of caveolin-1 in smooth muscle cells. This caveolin-1 in smooth muscle cells switches from being an antiproliferative factor to a proproliferative one and participates in cell proliferation and cell migration, possibly leading to irreversible PH. In contrast, the disruption of endothelial caveolin-1 is not observed in the hypoxia-induced PH, a reversible form of PH. However, proliferative pathways are activated in this model, indicating caveolin-1 dysfunction. Thus disruption or dysfunction of endothelial caveolin-1 leads to PH, and the status of caveolin-1 may determine the reversibility versus irreversibility of PH. This article reviews the role of caveolin-1 and cell membrane integrity in the pathogenesis and progression of PH.
Collapse
Affiliation(s)
- Rajamma Mathew
- Section of Pediatric Cardiology and Department of Physiology, Maria Fareri Children's Hospital/New York Medical College, Valhalla, New York
| |
Collapse
|
27
|
Maurya MR, Gupta S, Li X, Fahy E, Dinasarapu AR, Sud M, Brown HA, Glass CK, Murphy RC, Russell DW, Dennis EA, Subramaniam S. Analysis of inflammatory and lipid metabolic networks across RAW264.7 and thioglycolate-elicited macrophages. J Lipid Res 2013; 54:2525-42. [PMID: 23776196 DOI: 10.1194/jlr.m040212] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Studies of macrophage biology have been significantly advanced by the availability of cell lines such as RAW264.7 cells. However, it is unclear how these cell lines differ from primary macrophages such as thioglycolate-elicited peritoneal macrophages (TGEMs). We used the inflammatory stimulus Kdo2-lipid A (KLA) to stimulate RAW264.7 and TGEM cells. Temporal changes of lipid and gene expression levels were concomitantly measured and a systems-level analysis was performed on the fold-change data. Here we present a comprehensive comparison between the two cell types. Upon KLA treatment, both RAW264.7 and TGEM cells show a strong inflammatory response. TGEM (primary) cells show a more rapid and intense inflammatory response relative to RAW264.7 cells. DNA levels (fold-change relative to control) are reduced in RAW264.7 cells, correlating with greater downregulation of cell cycle genes. The transcriptional response suggests that the cholesterol de novo synthesis increases considerably in RAW264.7 cells, but 25-hydroxycholesterol increases considerably in TGEM cells. Overall, while RAW264.7 cells behave similarly to TGEM cells in some ways and can be used as a good model for inflammation- and immune function-related kinetic studies, they behave differently than TGEM cells in other aspects of lipid metabolism and phenotypes used as models for various disorders such as atherosclerosis.
Collapse
Affiliation(s)
- Mano R Maurya
- Department of Bioengineering, University of California at San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Zhang PX, Murray TS, Villella VR, Ferrari E, Esposito S, D'Souza A, Raia V, Maiuri L, Krause DS, Egan ME, Bruscia EM. Reduced caveolin-1 promotes hyperinflammation due to abnormal heme oxygenase-1 localization in lipopolysaccharide-challenged macrophages with dysfunctional cystic fibrosis transmembrane conductance regulator. THE JOURNAL OF IMMUNOLOGY 2013; 190:5196-206. [PMID: 23606537 DOI: 10.4049/jimmunol.1201607] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We have previously reported that TLR4 signaling is increased in LPS-stimulated cystic fibrosis (CF) macrophages (MΦs), contributing to the robust production of proinflammatory cytokines. The heme oxygenase-1 (HO-1)/CO pathway modulates cellular redox status, inflammatory responses, and cell survival. The HO-1 enzyme, together with the scaffold protein caveolin 1 (CAV-1), also acts as a negative regulator of TLR4 signaling in MΦs. In this study, we demonstrate that in LPS-challenged CF MΦs, HO-1 does not compartmentalize normally to the cell surface and instead accumulates intracellularly. The abnormal HO-1 localization in CF MΦs in response to LPS is due to decreased CAV-1 expression, which is controlled by the cellular oxidative state, and is required for HO-1 delivery to the cell surface. Overexpression of HO-1 or stimulating the pathway with CO-releasing molecules enhances CAV-1 expression in CF MΦs, suggesting a positive-feed forward loop between HO-1/CO induction and CAV-1 expression. These manipulations re-established HO-1 and CAV-1 cell surface localization in CF MΦs. Consistent with restoration of HO-1/CAV-1-negative regulation of TLR4 signaling, genetic or pharmacological (CO-releasing molecule 2) induced enhancement of this pathway decreased the inflammatory response of CF MΦs and CF mice treated with LPS. In conclusion, our results demonstrate that the counterregulatory HO-1/CO pathway, which is critical in balancing and limiting the inflammatory response, is defective in CF MΦs through a CAV-1-dependent mechanism, exacerbating the CF MΦ response to LPS. This pathway could be a potential target for therapeutic intervention for CF lung disease.
Collapse
Affiliation(s)
- Ping-Xia Zhang
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06509, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Basu Roy UK, Henkhaus RS, Loupakis F, Cremolini C, Gerner EW, Ignatenko NA. Caveolin-1 is a novel regulator of K-RAS-dependent migration in colon carcinogenesis. Int J Cancer 2013; 133:43-57. [PMID: 23280667 DOI: 10.1002/ijc.28001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 11/12/2012] [Accepted: 12/05/2012] [Indexed: 12/13/2022]
Abstract
Caveolin-1 is an essential component of membrane caveolae. It is an important regulator of cellular processes such as signal transduction and endocytosis. We report here, for the first time, that caveolin-1 is a target of the K-RAS oncogene in colon carcinogenesis. Caveolin-1 is induced in colon cancer cells and in human colon tumor samples, in response to K-RAS activating mutations. An activated K-RAS oncogene transcriptionally induces caveolin-1 expression in human colon cancer cells and this effect is not restricted to the type of activating K-RAS mutation. Inhibition of the P-I3 Kinase-AKT pathway, but not the ERK MAPK pathway, both important K-RAS effectors, leads to a decrease in caveolin-1 expression indicating that the AKT pathway is involved in caveolin-1 expression in response to an activated K-RAS. Increased AKT signaling induces caveolin-1 expression by increasing the activity of the transcription factor, Sp1. Interestingly; caveolin-1 depletion alters K-RAS-dependent signaling by decreasing Grb2-SOS activity. Consistent with these finding, caveolin-1-depleted cells shows decreased migration in vitro. However, caveolin-1 overexpression by itself does not increase migration whereas an activated Src can increase migration in a caveolin-1-dependent manner. This increased migration is highly dependent on the RhoA GTPase, indicating that an activated K-RAS modulates migration in part via caveolin-1 induction, and increasing RhoA activity via phospho-caveolin-1. Our findings indicate that K-RAS regulates both caveolin-1 expression and other factors affecting caveolin-1 functions in colon cancer-derived cell migration.
Collapse
Affiliation(s)
- Upal K Basu Roy
- Department of Biochemistry and Molecular Biophysics, Biochemistry and Molecular and Cellular Biology Graduate Program, University of Arizona, Tucson, AZ, USA
| | | | | | | | | | | |
Collapse
|
30
|
Cav1 suppresses tumor growth and metastasis in a murine model of cutaneous SCC through modulation of MAPK/AP-1 activation. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 182:992-1004. [PMID: 23267770 DOI: 10.1016/j.ajpath.2012.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 10/03/2012] [Accepted: 11/08/2012] [Indexed: 01/17/2023]
Abstract
Caveolin-1 (Cav1) is a scaffolding protein that serves to regulate the activity of several signaling molecules. Its loss has been implicated in the pathogenesis of several types of cancer, but its role in the development and progression of cutaneous squamous cell carcinoma (cSCC) remains largely unexplored. Herein, we use the keratinocyte cell line PAM212, a murine model of cSCC, to determine the function of Cav1 in skin tumor biology. We first show that Cav1 overexpression decreases cell and tumor growth, whereas Cav1 knockdown increases these attributes in PAM212 cells. In addition, Cav1 knockdown increases the invasive ability and incidence of spontaneous lymph node metastasis. Finally, we demonstrate that Cav1 knockdown increases extracellular signaling-related kinase 1/2 mitogen-activated protein kinase/activator protein-1 pathway activation. We attribute the growth and invasive advantage conferred by Cav1 knockdown to increased expression of activator protein-1 transcriptional targets, including cyclin D1 and keratin 18, which show inverse expression in PAM212 based on the expression level of Cav1. In summary, we demonstrate that loss of Cav1 affects several characteristics associated with aggressive human skin tumors and that this protein may be an important modulator of tumor growth and invasion in cSCC.
Collapse
|
31
|
Lal H, Verma SK, Feng H, Golden HB, Gerilechaogetu F, Nizamutdinov D, Foster DM, Glaser SS, Dostal DE. Caveolin and β1-integrin coordinate angiotensinogen expression in cardiac myocytes. Int J Cardiol 2012; 168:436-45. [PMID: 23058350 DOI: 10.1016/j.ijcard.2012.09.131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 06/23/2012] [Accepted: 09/22/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND The cardiac renin-angiotensin system (RAS) has been implicated in mediating myocyte hypertrophy and remodeling, although the biochemical mechanisms responsible for regulating the local RAS are poorly understood. Caveolin-1 (Cav-1)/Cav-3 double-knockout mice display cardiac hypertrophy, and in vitro disruption of lipid rafts/caveolae using methyl-β-cyclodextrin (MβCD) abolishes cardiac protection. METHODS In this study, neonatal rat ventricular myocytes (NRVM) were used to determine whether lipid rafts/caveolae may be involved in the regulation of angiotensinogen (Ao) gene expression, a substrate of the RAS system. RESULTS Treatment with MβCD caused a time-dependent upregulation of Ao gene expression, which was associated with differential regulation of mitogen-activated protein (MAP) kinases ERK1/2, p38 and JNK phosphorylation. JNK was highly phosphorylated shortly after MβCD treatment (2-30 min), whereas marked activation of ERK1/2 and p38 occurred much later (2-4h). β1D-Integrin was required for MβCD-induced activation of the MAP kinases. Pharmacologic inhibition of ERK1/2 and JNK enhanced MβCD-induced Ao gene expression, whereas p38 blockade inhibited this response. Adenovirus-mediated expression of wild-type p38α enhanced MβCD-induced Ao gene expression; conversely expression of dominant negative p38α blocked the stimulatory effects of MβCD. Expression of Cav-3 siRNA stimulated Ao gene expression, whereas overexpression of Cav-3 was inhibitory. Cav-1 and Cav-3 expression levels were found to be positively regulated by p38, but unaffected by ERK1/2 and JNK. CONCLUSION Collectively, these studies indicate that lipid rafts/caveolae couple to Ao gene expression through a mechanism that involves β1-integrin and the differential actions of MAP kinase family members.
Collapse
Affiliation(s)
- Hind Lal
- Center for Translational Medicine, Temple University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ma WY, Zhuang L, Cai DX, Zhong H, Zhao C, Sun Q. Inverse Correlation between Caveolin-1 Expression and Clinical Severity in Psoriasis Vulgaris. J Int Med Res 2012. [PMID: 23206456 DOI: 10.1177/030006051204000513] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Objective: This study investigated the clinical significance of expression of caveolin-1 - a plasma membrane protein involved in caveola formation, endocytosis, signal transduction and angiogenesis - inthe pathogenesis of psoriasis vulgaris. Methods: A total of 20 patients with psoriasis vulgaris and 20 healthy volunteers were recruited. The expressions of caveolin-1, Ki-67 (marker of cell proliferation) and CD34 (marker of angiogenesis) in skin biopsies were detected by immunohistochemistry, and the level of caveolin-1 protein was quantified by Western blotting. Clinical severity was assessed using the Psoriasis Area and Severity Index (PASI) score. Correlations between caveolin-1 expression and psoriasis severity, cell proliferation and angiogenesis were analysed using the Spearman rank correlation test. Results: Expression of caveolin-1 was significantly lower in psoriasis samples than in healthy skin samples. In psoriasis lesions, the level of caveolin-1 expression was inversely correlated with the severity of psoriasis, cell proliferation and angiogenesis. Conclusions: The level of caveolin-1 expression seems to be related to the clinical severity of psoriasis, and may play a role in the abnormal keratinocyte hyperplasia and angiogenesis seen in this condition.
Collapse
Affiliation(s)
- WY Ma
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, China
| | - L Zhuang
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, China
| | - DX Cai
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, China
| | - H Zhong
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, China
| | - C Zhao
- School of Medicine, Shandong University, Jinan, China
| | - Q Sun
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
33
|
Arreche ND, Sarati LI, Martinez CR, Fellet AL, Balaszczuk AM. Contribution of caveolin-1 to ventricular nitric oxide in age-related adaptation to hypovolemic state. ACTA ACUST UNITED AC 2012; 179:43-9. [PMID: 22954805 DOI: 10.1016/j.regpep.2012.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2011] [Revised: 06/24/2012] [Accepted: 08/27/2012] [Indexed: 11/26/2022]
Abstract
Our previous results have shown that hypovolemic state induced by acute hemorrhage in young anesthetized rats triggers heterogeneous and dynamic nitric oxide synthase (NOS) activation, modulating the cardiovascular response. Involvement of the nitric oxide pathway is both isoform-specific and time-dependent. The aim of the present study was to investigate changes in activity and protein levels of the different NOS forms, changes in the abundance of caveolin-1 during hypovolemic state and caveolin-1/eNOS association using young and middle-aged rats. Therefore, we studied (i) changes in NOS activity and protein levels and (ii) caveolin-1 abundance, as well as its association with endothelial NOS (eNOS) in ventricles from young and middle-aged rats during hypovolemic state. We used 2-month (young) and 12-month (middle-aged) old male Sprague-Dawley rats. Animals were divided into two groups (n=14/group): (a) sham; (b) hemorrhaged animals (20% blood loss). With advancing age, we observed an increase in ventricle NOS activity accompanied by a decrease in eNOS and caveolin-1 protein levels, but increased inducible NOS (iNOS). We also observed that aging is associated with caveolin-1 dissociation from eNOS. Myocardia from young and middle-aged rats subjected to hemorrhage-induced hypovolemia exhibited an increase in NOS activity and protein levels with a reduction in caveolin-1 abundance, accompanied by a greater dissociation between eNOS and its regulatory protein. Further, an increase in iNOS protein levels after blood loss was observed only in middle-aged rats. Our evidence suggests that aging and acute hemorrhage contribute to the development of upregulation in NOS activity. Our findings demonstrate that specific expression patterns of ventricular NOS isoforms, alterations in the amount of caveolin-1 and caveolin-1/eNOS interaction are involved in aged-related adjustment to hypovolemic state.
Collapse
Affiliation(s)
- Noelia D Arreche
- Department of Physiology, School of Pharmacy and Biochemistry, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | | | | | | | | |
Collapse
|
34
|
Yang XY, Huang CC, Kan QM, Li Y, Liu D, Zhang XC, Sato T, Yamagata S, Yamagata T. Calcium regulates caveolin-1 expression at the transcriptional level. Biochem Biophys Res Commun 2012; 426:334-41. [PMID: 22940132 DOI: 10.1016/j.bbrc.2012.08.079] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 08/15/2012] [Indexed: 11/29/2022]
Abstract
Caveolin-1, an indispensable component of caveolae serving as a transformation suppressor protein, is highly expressed in poorly metastatic mouse osteosarcoma FBJ-S1 cells while highly metastatic FBJ-LL cells express low levels of caveolin-1. Calcium concentration is higher in FBJ-S1 cells than in FBJ-LL cells; therefore, we investigated the possibility that calcium signaling positively regulates caveolin-1 in mouse FBJ-S1 cells. When cells were treated with the calcium channel blocker nifedipine, cyclosporin A (a calcineurin inhibitor), or INCA-6 (a nuclear factor of activated T-cells [NFAT] inhibitor), caveolin-1 expression at the mRNA and protein levels decreased. RNA silencing of voltage-dependent L-type calcium channel subunit alpha-1C resulted in suppression of caveolin-1 expression. This novel caveolin-1 regulation pathway was also identified in mouse NIH 3T3 cells and Lewis lung carcinoma cells. These results indicate that caveolin-1 is positively regulated at the transcriptional level through a novel calcium signaling pathway mediated by L-type calcium channel/Ca(2+)/calcineurin/NFAT.
Collapse
Affiliation(s)
- Xiao-Yan Yang
- Laboratory of Tumor Biology and Glycobiology, Department of Life Sciences, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sonnino S, Prioni S, Chigorno V, Prinetti A. Interactions Between Caveolin-1 and Sphingolipids, and Their Functional Relevance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 749:97-115. [DOI: 10.1007/978-1-4614-3381-1_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
36
|
Huang C, Qiu Z, Wang L, Peng Z, Jia Z, Logsdon CD, Le X, Wei D, Huang S, Xie K. A novel FoxM1-caveolin signaling pathway promotes pancreatic cancer invasion and metastasis. Cancer Res 2011; 72:655-65. [PMID: 22194465 DOI: 10.1158/0008-5472.can-11-3102] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Caveolin-1 (Cav-1), a principal structural component of caveolar membrane domains, contributes to cancer development but its precise functional roles and regulation remain unclear. In this study, we determined the oncogenic function of Cav-1 in preclinical models of pancreatic cancer and in human tissue specimens. Cav-1 expression levels correlated with metastatic potential and epithelial-mesenchymal transition (EMT) in both mouse and human pancreatic cancer cells. Elevated levels in cells promoted EMT, migration, invasion, and metastasis in animal models, whereas RNA interference (RNAi)-mediated knockdown inhibited these processes. We determined that levels of Cav-1 and the Forkhead transcription factor FoxM1 correlated directly in pancreatic cancer cells and tumor tissues. Enforced expression of FoxM1 increased Cav-1 levels, whereas RNAi-mediated knockdown of FoxM1 had the opposite effect. FoxM1 directly bound to the promoter region of Cav-1 gene and positively transactivated its activity. Collectively, our findings defined Cav-1 as an important downstream oncogenic target of FoxM1, suggesting that dysregulated signaling of this novel FoxM1-Cav-1 pathway promotes pancreatic cancer development and progression.
Collapse
Affiliation(s)
- Chen Huang
- Shanghai Key Laboratory of Pancreatic Diseases Research, Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Prinetti A, Cao T, Illuzzi G, Prioni S, Aureli M, Gagliano N, Tredici G, Rodriguez-Menendez V, Chigorno V, Sonnino S. A glycosphingolipid/caveolin-1 signaling complex inhibits motility of human ovarian carcinoma cells. J Biol Chem 2011; 286:40900-10. [PMID: 21949119 DOI: 10.1074/jbc.m111.286146] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The genetic (stable overexpression of sialyltransferase I, GM3 synthase) or pharmacological (selective pressure by N-(4-hydroxyphenyl)retinamide)) manipulation of A2780 human ovarian cancer cells allowed us to obtain clones characterized by higher GM3 synthase activity compared with wild-type cells. Clones with high GM3 synthase expression had elevated ganglioside levels, reduced in vitro cell motility, and enhanced expression of the membrane adaptor protein caveolin-1 with respect to wild-type cells. In high GM3 synthase-expressing clones, both depletion of gangliosides by treatment with the glucosylceramide synthase inhibitor D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol and silencing of caveolin-1 by siRNA were able to strongly increase in vitro cell motility. The motility of wild-type, low GM3 synthase-expressing cells was reduced in the presence of a Src inhibitor, and treatment of these cells with exogenous gangliosides, able to reduce their in vitro motility, inactivated c-Src kinase. Conversely, ganglioside depletion by D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol treatment or caveolin-1 silencing in high GM3 synthase-expressing cells led to c-Src kinase activation. In high GM3 synthase-expressing cells, caveolin-1 was associated with sphingolipids, integrin receptor subunits, p130(CAS), and c-Src forming a Triton X-100-insoluble noncaveolar signaling complex. These data suggest a role for gangliosides in regulating tumor cell motility by affecting the function of a signaling complex organized by caveolin-1, responsible for Src inactivation downstream to integrin receptors, and imply that GM3 synthase is a key target for the regulation of cell motility in human ovarian carcinoma.
Collapse
Affiliation(s)
- Alessandro Prinetti
- Department of Medical Chemistry, Biochemistry and Biotechnology, University of Milan, 20090 Segrate, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Mulas MF, Abete C, Pulisci D, Pani A, Massidda B, Dessì S, Mandas A. Cholesterol esters as growth regulators of lymphocytic leukaemia cells. Cell Prolif 2011; 44:360-71. [PMID: 21645151 DOI: 10.1111/j.1365-2184.2011.00758.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Alterations in plasma lipid profile and in intracellular cholesterol homoeostasis have been described in various malignancies; however, significance of these alterations, if any, in cancer biology is not clear. The aim of the present study was to investigate a possible correlation between alterations in cholesterol metabolism and expansion of leukaemia cell numbers. MATERIALS AND METHODS Lipid profiles in plasma and in primary leukaemia cells isolated from patients with acute or chronic lymphocytic leukaemia (ALL and CLL) were studied. RESULTS AND CONCLUSIONS Decreased levels of HDL-C were observed in plasma of leukaemic patients, levels of total cholesterol, LDL-C, triglycerides and phospholipids were unchanged or only slightly increased. As compared to normal lymphocytes, freshly isolated leukaemic cells showed increased levels of cholesterol esters and reduction in free cholesterol. Growth stimulation of ALL and CLL cells with phytohemagglutinin led to further increase in levels of cholesterol esters. Conversely, treatment with an inhibitor of cell proliferation such as the mTOR inhibitor, RAD, caused decline in population growth rate of leukaemia cells, which was preceded by sharp reduction in rate of cholesterol esterification. On the other hand, exposure of leukaemic cells to two inhibitors of cholesterol esterification, progesterone and SaH 58-035, caused 60% reduction in their proliferation rate. In addition to demonstrating tight correlation between cell number expansion and cholesterol esterification in leukaemic cells, these results suggest that pathways that control cholesterol esterification might represent a promising targets for novel anticancer strategies.
Collapse
Affiliation(s)
- M F Mulas
- Department of Internal Medicine, University of Cagliari, Italy
| | | | | | | | | | | | | |
Collapse
|
39
|
Chen W, Chen Y, Qin L, Li A, Zhao X, Wang X, Xiao S, Mo D. Transcription factor Sp1 is essential for the regulation of the porcine caveolin-1 gene. DNA Cell Biol 2011; 30:491-7. [PMID: 21631282 DOI: 10.1089/dna.2010.1202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Caveolin-1 (CAV-1) is a key structural component of caveolae that regulates cholesterol. Employing transgenic techniques to regulate the cholesterol content of pork through CAV-1 is hindered by our lack of knowledge about its regulation. To investigate the regulatory mechanism of porcine CAV-1, a DNA segment containing the 5'-flanking region of CAV-1 was isolated from porcine genomic DNA and sequenced. The luciferase reporter assay detected five cis-acting elements for efficient expression of the CAV-1 gene at the region spanning nucleotides -213 to -20 with serially deleted 5'-flanking sequences and site-directed mutants, -123 to -114 was the core promoter. The electrophoretic mobility shift assay demonstrated potential binding of Sp1 protein to this core promoter. The purpose of this study is to systematically elucidate the transcriptional regulation mechanism of porcine CAV-1 and to contribute to the investigation of the interaction between CAV-1 and cholesterol.
Collapse
Affiliation(s)
- Weiquan Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Chiu WT, Lee HT, Huang FJ, Aldape KD, Yao J, Steeg PS, Chou CY, Lu Z, Xie K, Huang S. Caveolin-1 upregulation mediates suppression of primary breast tumor growth and brain metastases by stat3 inhibition. Cancer Res 2011; 71:4932-43. [PMID: 21622714 DOI: 10.1158/0008-5472.can-10-4249] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Stat3 activation has been implicated as an important driver of brain metastasis in breast cancer, but the critical targets of Stat3 in this process are yet to be fully defined. In this study, we identified the lipid raft organizing protein Caveolin-1 (Cav-1) as a critical genetic target of Stat3 in this process. In human breast cancers, we found that activated Stat3 correlated with attenuation of Cav-1 in brain metastases relative to primary tumors. Cav-1 promoter activity and gene expression were increased by overexpressing an activated form of Stat3 but decreased by attenuation of Stat3 activity or expression. We identified putative Stat3-binding elements in the Cav-1 promoter and showed a direct repression of Cav-1 transcription by Stat3. Reciprocally, we showed that strategies to increase or decrease Cav-1 expression were sufficient to attenuate or promote breast cancer cell invasion. Furthermore, increased expression of Cav-1 phenocopied the effects of Stat3 activation in blocking primary tumor growth and abrogating formation of brain metastases. Collectively, our findings provide clinical and mechanistic evidence that Cav-1 is a critical target for suppression by Stat3 in driving invasion and metastasis of breast cancer cells.
Collapse
Affiliation(s)
- Wen-Tai Chiu
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Cell-specific dual role of caveolin-1 in pulmonary hypertension. Pulm Med 2011; 2011:573432. [PMID: 21660237 PMCID: PMC3109422 DOI: 10.1155/2011/573432] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Accepted: 03/10/2011] [Indexed: 12/15/2022] Open
Abstract
A wide variety of cardiopulmonary and systemic diseases are known to lead to pulmonary hypertension (PH). A number of signaling pathways have been implicated in PH; however, the precise mechanism/s leading to PH is not yet clearly understood. Caveolin-1, a membrane scaffolding protein found in a number of cells including endothelial and smooth muscle cells, has been implicated in PH. Loss of endothelial caveolin-1 is reported in clinical and experimental forms of PH. Caveolin-1, also known as a tumor-suppressor factor, interacts with a number of transducing molecules that reside in or are recruited to caveolae, and it inhibits cell proliferative pathways. Not surprisingly, the rescue of endothelial caveolin-1 has been found not only to inhibit the activation of proliferative pathways but also to attenuate PH. Recently, it has emerged that during the progression of PH, enhanced expression of caveolin-1 occurs in smooth muscle cells, where it facilitates cell proliferation, thus contributing to worsening of the disease. This paper summarizes the cell-specific dual role of caveolin-1 in PH.
Collapse
|
42
|
Liu XD, Chen HB, Tong Q, Li XY, Zhu MJ, Wu ZF, Zhou R, Zhao SH. Molecular characterization of caveolin-1 in pigs infected with Haemophilus parasuis. THE JOURNAL OF IMMUNOLOGY 2011; 186:3031-46. [PMID: 21282513 DOI: 10.4049/jimmunol.0902687] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Caveolin-1 (Cav1) plays a critical role in the invasion of pathogenic microbes into host cells, yet little is known about porcine Cav1. In this study, we provide the molecular characterization of Cav1 in pigs following stimulation with LPS/polyinosinic-polycytidylic acid as well as during infection with Haemophilus parasuis. The porcine Cav1 gene is 35 kb long and is located at SSC18q21; two isoforms (Cav1-α and Cav1-β) are produced by alternative splicing. Three point mutations were identified in the coding region of the gene, two of which were significantly associated with nine immunological parameters in Landrace pigs, including the Ab response against porcine reproductive and respiratory syndrome virus and lymphocyte counts. Promoter analysis indicated that NF-κB activates both Cav1 transcripts, but the forkhead gene family specifically regulates Cav1-β in the pig. Porcine Cav1 is expressed ubiquitously, with Cav1-α more abundantly expressed than Cav1-β in all tissues investigated. Basal expression levels of Cav1 in PBMCs are relatively similar across different pig breeds. LPS and polyinosinic-polycytidylic acid markedly induced the expression of Cav1 in porcine kidney-15 cells in vitro, likely through NF-κB activation. Pigs infected with H. parasuis exhibited decreased expression of Cav1, particularly in seriously impaired organs such as the brain. This study provides new evidence that supports the use of Cav1 as a potential diagnostic and genetic marker for disease resistance in animal breeding. In addition, our results suggest that Cav1 may be implicated in the pathogenesis of Glasser's disease, which is caused by H. parasuis.
Collapse
Affiliation(s)
- Xiang-Dong Liu
- Key Laboratory of Agricultural Animal Genetics and Breeding, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Prinetti A, Prioni S, Loberto N, Aureli M, Nocco V, Illuzzi G, Mauri L, Valsecchi M, Chigorno V, Sonnino S. Aberrant glycosphingolipid expression and membrane organization in tumor cells: consequences on tumor-host interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 705:643-67. [PMID: 21618134 DOI: 10.1007/978-1-4419-7877-6_34] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Alessandro Prinetti
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Via Fratelli Cervi 93, 20090 Segrate, Milano, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Tomasi V. Signal transduction in neurons: effects of cellular prion protein on fyn kinase and ERK1/2 kinase. IMMUNITY & AGEING 2010; 7 Suppl 1:S5. [PMID: 21172064 PMCID: PMC3024879 DOI: 10.1186/1742-4933-7-s1-s5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background It has been reported that cellular prion protein (PrPc) co-localizes with caveolin-1 and participates to signal transduction events by recruiting Fyn kinase. As PrPc is a secreted protein anchored to the outer surface membrane through a glycosylphosphatidylinositol (GPI) anchor (secPrP) and caveolin-1 is located in the inner leaflet of plasma membrane, there is a problem of how the two proteins can physically interact each other and transduce signals. Results By using the GST-fusion proteins system we observed that PrPc strongly interacts with caveolin-1 scaffolding domain and with a caveolin-1 hydrophilic C-terminal region, but not with the caveolin-1 N-terminal region. In vitro binding experiments were also performed to define the site(s) of PrPc interacting with cav-1. The results are consistent with a participation of PrPc octapeptide repeats motif in the binding to caveolin-1 scaffolding domain. The caveolar localization of PrPc was ascertained by co-immunoprecipitation, by co-localization after flotation in density gradients and by confocal microscopy analysis of PrPc and caveolin-1 distributions in a neuronal cell line (GN11) expressing caveolin-1 at high levels. Conclusions We observed that, after antibody-mediated cross-linking or copper treatment, PrPc was internalized probably into caveolae. We propose that following translocation from rafts to caveolae or caveolae-like domains, secPrP could interact with caveolin-1 and induce signal transduction events.
Collapse
Affiliation(s)
- Vittorio Tomasi
- Department of Experimental Biology, University of Bologna Via Selmi, 3 40126 - Bologna, Italy.
| |
Collapse
|
45
|
Yudoh K, Shi Y, Karasawa R. Angiogenic growth factors inhibit chondrocyte ageing in osteoarthritis: potential involvement of catabolic stress-induced overexpression of caveolin-1 in cellular ageing. Int J Rheum Dis 2010; 12:90-9. [PMID: 20374325 DOI: 10.1111/j.1756-185x.2009.01390.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Recently, attention has been attracted by the finding that overexpression of caveolin-1 induces cellular senescence in age-related diseases. We aimed to ascertain whether angiogenic growth factors (AGFs) can inhibit interleukin (IL)-1beta-induced senescence in human chondrocytes by downregulation of caveolin-1. METHODS We investigated the intracellular signalling pathways involved in chondrocyte ageing. Human chondrocytes were isolated from the articular cartilage of patients undergoing arthroplastic knee surgery in osteoarthritis (OA). Chondrocytes were stimulated with or without IL-1beta (10 ng/mL) in the presence or absence of vascular endothelial growth factor, basic fibroblast growth factor or hepatocyte growth factor (20 ng/mL). After 72-h incubation, we observed the expression of caveolin-1 in human chondrocytes by immunohistochemistry, and analysed the protein levels of caveolin-1 by Western blot. We examined the time-course of phosphorylation patterns of mammalian mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3-K) by Western blot, and used several specific protein kinase inhibitors to evaluate the involvement of the intracellular signalling pathways. Also, chondrocyte replicative lifespan was analyzed in the presence or absence of AGFs. RESULTS Treatment with AGFs inhibited IL-1beta-induced overexpression of caveolin-1 in human OA chondrocytes. Treatment with AGFs all down-regulated protein levels of IL-1beta-accelerated expression of caveolin-1 in chondrocytes. IL-1beta significantly decreased the cellular replicative lifespan in chondrocytes. Treatment with AGFs prevented the IL-1beta-induced shortening of chondrocyte replicative lifespan. The specific inhibitors for MAPK/extracellular signal-regulated kinase and PI3-K cancelled the AGF-induced downregulation of overexpression of caveolin-1. CONCLUSION Our results suggest that AGFs downregulated IL-1beta-induced chondrocyte ageing and overexpression of caveolin-1 in human chondrocytes, which is mediated by kinase cascades involving the p42/44 MAP kinase and PI3-K/Akt signalling pathways.
Collapse
Affiliation(s)
- Kazuo Yudoh
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Japan.
| | | | | |
Collapse
|
46
|
Kozak LP, Newman S, Chao PM, Mendoza T, Koza RA. The early nutritional environment of mice determines the capacity for adipose tissue expansion by modulating genes of caveolae structure. PLoS One 2010; 5:e11015. [PMID: 20574519 PMCID: PMC2888576 DOI: 10.1371/journal.pone.0011015] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 05/14/2010] [Indexed: 01/11/2023] Open
Abstract
While the phenomenon linking the early nutritional environment to disease susceptibility exists in many mammalian species, the underlying mechanisms are unknown. We hypothesized that nutritional programming is a variable quantitative state of gene expression, fixed by the state of energy balance in the neonate, that waxes and wanes in the adult animal in response to changes in energy balance. We tested this hypothesis with an experiment, based upon global gene expression, to identify networks of genes in which expression patterns in inguinal fat of mice have been altered by the nutritional environment during early post-natal development. The effects of over- and under-nutrition on adiposity and gene expression phenotypes were assessed at 5, 10, 21 days of age and in adult C57Bl/6J mice fed chow followed by high fat diet for 8 weeks. Under-nutrition severely suppressed plasma insulin and leptin during lactation and diet-induced obesity in adult mice, whereas over-nourished mice were phenotypically indistinguishable from those on a control diet. Food intake was not affected by under- or over-nutrition. Microarray gene expression data revealed a major class of genes encoding proteins of the caveolae and cytoskeleton, including Cav1, Cav2, Ptrf (Cavin1), Ldlr, Vldlr and Mest, that were highly associated with adipose tissue expansion in 10 day-old mice during the dynamic phase of inguinal fat development and in adult animals exposed to an obesogenic environment. In conclusion gene expression profiles, fat mass and adipocyte size in 10 day old mice predicted similar phenotypes in adult mice with variable diet-induced obesity. These results are supported by phenotypes of KO mice and suggest that when an animal enters a state of positive energy balance adipose tissue expansion is initiated by coordinate changes in mRNA levels for proteins required for modulating the structure of the caveolae to maximize the capacity of the adipocyte for lipid storage.
Collapse
Affiliation(s)
- Leslie P Kozak
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA.
| | | | | | | | | |
Collapse
|
47
|
Toni M, Spisni E, Griffoni C, Santi S, Riccio M, Lenaz P, Tomasi V. Cellular prion protein and caveolin-1 interaction in a neuronal cell line precedes Fyn/Erk 1/2 signal transduction. J Biomed Biotechnol 2010; 2006:69469. [PMID: 17489019 PMCID: PMC1559926 DOI: 10.1155/jbb/2006/69469] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It has been reported that cellular prion protein (PrPc) is enriched in caveolae or caveolae-like domains with caveolin-1 (Cav-1)
participating to signal transduction events by Fyn kinase recruitment. By using the Glutathione-S-transferase (GST)-fusion proteins
assay, we observed that PrPc strongly interacts in vitro with Cav-1. Thus, we ascertained the PrPc caveolar localization in a
hypothalamic neuronal cell line (GN11), by confocal microscopy analysis, flotation on density gradient, and coimmunoprecipitation
experiments. Following the anti-PrPc antibody-mediated stimulation of live GN11 cells, we observed that PrPc clustered on
plasma membrane domains rich in Cav-1 in which Fyn kinase converged to be activated. After these events, a signaling cascade
through p42/44 MAP kinase (Erk 1/2) was triggered, suggesting that following translocations from rafts to caveolae or caveolaelike
domains PrPc could interact with Cav-1 and induce signal transduction events.
Collapse
Affiliation(s)
- Mattia Toni
- Department of Experimental Biology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Enzo Spisni
- Department of Experimental Biology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Cristiana Griffoni
- Department of Experimental Biology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Spartaco Santi
- National Research Council, Institute of Cytomorphology, 40136 Bologna, Italy
| | - Massimo Riccio
- National Research Council, Institute of Cytomorphology, 40136 Bologna, Italy
| | - Patrizia Lenaz
- Department of Experimental Biology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Vittorio Tomasi
- Department of Experimental Biology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| |
Collapse
|
48
|
Integrin alpha1beta1 regulates epidermal growth factor receptor activation by controlling peroxisome proliferator-activated receptor gamma-dependent caveolin-1 expression. Mol Cell Biol 2010; 30:3048-58. [PMID: 20368353 DOI: 10.1128/mcb.00892-09] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Integrin alpha1beta1 negatively regulates the generation of profibrotic reactive oxygen species (ROS) by inhibiting epidermal growth factor receptor (EGFR) activation; however, the mechanism by which it does this is unknown. In this study, we show that caveolin-1 (Cav-1), a scaffolding protein that binds integrins and controls growth factor receptor signaling, participates in integrin alpha1beta1-mediated EGFR activation. Integrin alpha1-null mesangial cells (MCs) have reduced Cav-1 levels, and reexpression of the integrin alpha1 subunit increases Cav-1 levels, decreases EGFR activation, and reduces ROS production. Downregulation of Cav-1 in wild-type MCs increases EGFR phosphorylation and ROS synthesis, while overexpression of Cav-1 in the integrin alpha1-null MCs decreases EGFR-mediated ROS production. We further show that integrin alpha1-null MCs have increased levels of activated extracellular signal-regulated kinase (ERK), which leads to reduced activation of peroxisome proliferator-activated receptor gamma (PPARgamma), a transcription factor that positively regulates Cav-1 expression. Moreover, activation of PPARgamma or inhibition of ERK increases Cav-1 levels in the integrin alpha1-null MCs. Finally, we show that glomeruli of integrin alpha1-null mice have reduced levels of Cav-1 and activated PPARgamma but increased levels of phosphorylated EGFR both at baseline and following injury. Thus, integrin alpha1beta1 negatively regulates EGFR activation by positively controlling Cav-1 levels, and the ERK/PPARgamma axis plays a key role in regulating integrin alpha1beta1-dependent Cav-1 expression and consequent EGFR-mediated ROS production.
Collapse
|
49
|
Xiong J, Liu J, Rayner S, Li Y, Chen S. Protein-protein interaction reveals synergistic discrimination of cancer phenotype. Cancer Inform 2010; 9:61-6. [PMID: 20458363 PMCID: PMC2865773 DOI: 10.4137/cin.s3899] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cancer is a disease associated with the deregulation of multiple gene networks. Microarray data has permitted researchers to identify gene panel markers for diagnosis or prognosis of cancer but these are not sufficient to make specific mechanistic assertions about phenotype switches. We propose a strategy to identify putative mechanisms of cancer phenotypes by protein-protein interactions (PPI). We first extracted the logic status of a PPI via the relative expression of the corresponding gene pair. The joint association of a gene pair on a cancer phenotype was calculated by entropy minimization and assessed using a support vector machine. A typical predictor is “If Src high-expression, and Cav-1 low-expression, then cancer.” We achieved 90% accuracy on test data with a majority of predictions associated with the MAPK pathway, focal adhesion, apoptosis and cell cycle. Our results can aid in the development of phenotype discrimination biomarkers and identification of putative therapeutic interference targets for drug development.
Collapse
Affiliation(s)
- Jianghui Xiong
- School of Computer Science, Wuhan University, Wuhan, P.R. China.
| | | | | | | | | |
Collapse
|
50
|
Mayoral R, Valverde ÁM, Llorente Izquierdo C, González-Rodríguez Á, Boscá L, Martín-Sanz P. Impairment of transforming growth factor beta signaling in caveolin-1-deficient hepatocytes: role in liver regeneration. J Biol Chem 2010; 285:3633-3642. [PMID: 19966340 PMCID: PMC2823504 DOI: 10.1074/jbc.m109.072900] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 12/04/2009] [Indexed: 02/05/2023] Open
Abstract
Caveolin-1 (Cav-1) is the main structural protein of caveolae and plays an important role in various cellular processes such as vesicular transport, cholesterol homeostasis, and signal transduction pathways. The expression and functional role of Cav-1 have been reported in liver and in hepatocyte cell lines, in human cirrhotic liver, and in hepatocellular carcinomas. Previous studies demonstrated that Cav-1 was dispensable for liver regeneration, because Cav-1(-/-) animals survived and fully regenerated liver function and size after partial hepatectomy. In this study, we have investigated the mechanisms by which the lack of Cav-1 accelerates liver regeneration after partial hepatectomy. The data show that transforming growth factor beta (TGF-beta) signaling is impaired in regenerating liver of Cav-1(-/-) mice and in hepatocytes derived from these animals. TGF-beta receptors I and II do not colocalize in the same membrane fraction in the hepatocytes derived from Cav-1(-/-) mice, as Smad2/3 signaling decreased in the absence of Cav-1 at the time that the transcriptional corepressor SnoN accumulates. Accordingly, the expression of TGF-beta target genes, such as plasminogen activator inhibitor-1, is decreased due to the presence of the high levels of SnoN. Moreover, hepatocyte growth factor inhibited TGF-beta signaling in the absence of Cav-1 by increasing SnoN expression. Taken together, these data might help to unravel why Cav-1-deficient mice exhibit an accelerated liver regeneration after partial hepatectomy and add new insights on the molecular mechanisms controlling the initial commitment to hepatocyte proliferation.
Collapse
Affiliation(s)
- Rafael Mayoral
- From the Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas, Arturo Duperier 4, 28029 Madrid; the Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Villarroel 170, 08036 Barcelona, and
| | - Ángela M Valverde
- From the Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas, Arturo Duperier 4, 28029 Madrid; the Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Mallorca 183, 08036 Barcelona, Spain
| | - Cristina Llorente Izquierdo
- From the Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas, Arturo Duperier 4, 28029 Madrid
| | - Águeda González-Rodríguez
- From the Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas, Arturo Duperier 4, 28029 Madrid; the Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Mallorca 183, 08036 Barcelona, Spain
| | - Lisardo Boscá
- From the Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas, Arturo Duperier 4, 28029 Madrid; the Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Villarroel 170, 08036 Barcelona, and
| | - Paloma Martín-Sanz
- From the Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas, Arturo Duperier 4, 28029 Madrid; the Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Villarroel 170, 08036 Barcelona, and.
| |
Collapse
|