51
|
Huang SS, Liu IH, Chen CL, Chang JM, Johnson FE, Huang JS. 7-Dehydrocholesterol (7-DHC), But Not Cholesterol, Causes Suppression of Canonical TGF-β Signaling and Is Likely Involved in the Development of Atherosclerotic Cardiovascular Disease (ASCVD). J Cell Biochem 2017; 118:1387-1400. [PMID: 27862220 PMCID: PMC6123222 DOI: 10.1002/jcb.25797] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 11/14/2016] [Indexed: 02/02/2023]
Abstract
For several decades, cholesterol has been thought to cause ASCVD. Limiting dietary cholesterol intake has been recommended to reduce the risk of the disease. However, several recent epidemiological studies do not support a relationship between dietary cholesterol and/or blood cholesterol and ASCVD. Consequently, the role of cholesterol in atherogenesis is now uncertain. Much evidence indicates that TGF-β, an anti-inflammatory cytokine, protects against ASCVD and that suppression of canonical TGF-β signaling (Smad2-dependent) is involved in atherogenesis. We had hypothesized that cholesterol causes ASCVD by suppressing canonical TGF-β signaling in vascular endothelium. To test this hypothesis, we determine the effects of cholesterol, 7-dehydrocholesterol (7-DHC; the biosynthetic precursor of cholesterol), and other sterols on canonical TGF-β signaling. We use Mv1Lu cells (a model cell system for studying TGF-β activity) stably expressing the Smad2-dependent luciferase reporter gene. We demonstrate that 7-DHC (but not cholesterol or other sterols) effectively suppresses the TGF-β-stimulated luciferase activity. We also demonstrate that 7-DHC suppresses TGF-β-stimulated luciferase activity by promoting lipid raft/caveolae formation and subsequently recruiting cell-surface TGF-β receptors from non-lipid raft microdomains to lipid rafts/caveolae where TGF-β receptors become inactive in transducing canonical signaling and undergo rapid degradation upon TGF-β binding. We determine this by cell-surface 125 I-TGF-β-cross-linking and sucrose density gradient ultracentrifugation. We further demonstrate that methyl-β-cyclodextrin (MβCD), a sterol-chelating agent, reverses 7-DHC-induced suppression of TGF-β-stimulated luciferase activity by extrusion of 7-DHC from resident lipid rafts/caveolae. These results suggest that 7-DHC, but not cholesterol, promotes lipid raft/caveolae formation, leading to suppression of canonical TGF-β signaling and atherogenesis. J. Cell. Biochem. 118: 1387-1400, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - I-Hua Liu
- Department of Pharmacology, Institute for Drug Evaluation Platform, Development Center for Biotechnology, Taipei, Taiwan
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-Sen University and Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University and Academia Sinica, Kaohsiung, Taiwan
| | - Jia-Ming Chang
- Department of Pharmacology, Institute for Drug Evaluation Platform, Development Center for Biotechnology, Taipei, Taiwan
| | - Frank E. Johnson
- Department of Surgery, Saint Louis University Medical Center, 3635 Vista Ave., St. Louis, Missouri 63110
| | - Jung San Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, 1100 S. Grand Blvd., St. Louis, Missouri 63104
| |
Collapse
|
52
|
Pentabromophenol suppresses TGF-β signaling by accelerating degradation of type II TGF-β receptors via caveolae-mediated endocytosis. Sci Rep 2017; 7:43206. [PMID: 28230093 PMCID: PMC5322341 DOI: 10.1038/srep43206] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 01/23/2017] [Indexed: 12/15/2022] Open
Abstract
Pentabromophenol (PBP), a brominated flame retardant (BFR), is widely used in various consumer products. BFRs exert adverse health effects such as neurotoxic and endocrine-disrupting effects. In this study, we found that PBP suppressed TGF-β response by accelerating the turnover rate of TGF-β receptors. PBP suppressed TGF-β-mediated cell migration, PAI-1 promoter-driven reporter gene activation, and Smad2/3 phosphorylation in various cell types. Furthermore, PBP abolished TGF-β-mediated repression of E-cadherin expression, in addition to the induction of vimentin expression and N-cadherin and fibronectin upregulation, thus blocking TGF-β-induced epithelial–mesenchymal transition in A549 and NMuMG cells. However, this inhibition was not observed with other congeners such as tribromophenol and triiodophenol. TGF-β superfamily members play key roles in regulating various biological processes including cell proliferation and migration as well as cancer development and progression. The results of this in vitro study provide a basis for studies on the detailed relationship between PBP and modulation of TGF-β signalling. Because PBP is similar to other BFRs such as polybrominated diphenyl ethers (PBDEs), additional laboratory and mechanistic studies should be performed to examine BFRs as potential risk factors for tumorigenesis and other TGF-β-related diseases.
Collapse
|
53
|
Development of a novel imaging agent using peptide-coated gold nanoparticles toward brain glioma stem cell marker CD133. Acta Biomater 2017; 47:182-192. [PMID: 27721007 DOI: 10.1016/j.actbio.2016.10.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 09/28/2016] [Accepted: 10/05/2016] [Indexed: 12/13/2022]
Abstract
CD133 is known as biomarker for glioblastoma (GBM) and also serves as a marker for cancer stem cells (CSCs), which carry out tumorigenesis and resist conventional therapeutics. The presence of CD133-presenting CSC is a one of the factors in maintenance of the tumorigenic potential of GBM. Thus, CD133 is a potential target for accurate diagnosis of GBM, which could improve its poor prognosis for patients when CSCs are present. Herein we designed a small peptide-based imaging agent with stimulus-responsive properties. A novel small peptide, CBP4, was screened by a phage display technique, and demonstrated binding to the target CD133 (ECD) comparable to that of an antibody. As a quencher, we used gold nanoparticles (GNPs); the targeting peptide was conjugated to GNPs with high efficiency. By means of a quenching effect, the peptide-coated GNP showed 'signal on-off' properties depending upon the presence of the target. In addition, the particles exhibited biocompatibility when localized in the cytosol. Thus, this study demonstrated that the peptide-coated GNPs can be utilized as an imaging agent for accurate diagnosis of GBM, and further as a drug carrier for therapeutic approaches. STATEMENT OF SIGNIFICANCE The diagnosis and determination of prognosis made by cancer stem cell markers could be a key strategy to eradicate cancer stem cells and cure the cancer. The significance of this study is the characterization of quenching-based signal on-off mechanism and showed that the active targeting via peptide can contribute to the design of a stimulus-responsive cellular imaging agent. Moreover, small peptide based nano complexation showed specific recognition of the target stem cell and internalized on cellular cyotosol with stimulus responsive fluorescence. With its novel biocompatibility, the strategy might be a promising tool for drug carrier systems able to measure and visualize the delivered efficiency at intracellular sites.
Collapse
|
54
|
Li Y, Monteiro-Riviere NA. Mechanisms of cell uptake, inflammatory potential and protein corona effects with gold nanoparticles. Nanomedicine (Lond) 2016; 11:3185-3203. [DOI: 10.2217/nnm-2016-0303] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Aim: To assess inflammation, cellular uptake and endocytic mechanisms of gold nanoparticles (AuNP) in human epidermal keratinocytes with and without a protein corona. Materials & methods: Human epidermal keratinocytes were exposed to 40 and 80 nm AuNP with lipoic acid, polyethylene glycol (PEG) and branched polyethyleneimine (BPEI) coatings with and without a protein corona up to 48 h. Inhibitors were selected to characterize endocytosis. Results & conclusion: BPEI-AuNP showed the greatest uptake, while PEG-AuNP had the least. Protein coronas decreased uptake and affected their mechanism. AuNP uptake was energy-dependent, except for 40 nm lipoic-AuNP. Most AuNP were internalized by clathrin and lipid raft-mediated endocytosis, except for 40 nm PEG was by raft/noncaveolae mediated endocytosis. Coronas inhibited caveolae-mediated-endocytosis with lipoic acid and BPEI-AuNP and altered 40 nm PEG-AuNP from raft/noncaveolae to clathrin. Inflammatory responses decreased with a plasma corona. Results suggest protein coronas significantly affect cellular uptake and inflammatory responses of AuNP.
Collapse
Affiliation(s)
- Yang Li
- Nanotechnology Innovation Center of Kansas State University (NICKS), Kansas State University, Manhattan, KS, USA
| | - Nancy A Monteiro-Riviere
- Nanotechnology Innovation Center of Kansas State University (NICKS), Kansas State University, Manhattan, KS, USA
| |
Collapse
|
55
|
Wu S, Majeed SR, Evans TM, Camus MD, Wong NML, Schollmeier Y, Park M, Muppidi JR, Reboldi A, Parham P, Cyster JG, Brodsky FM. Clathrin light chains' role in selective endocytosis influences antibody isotype switching. Proc Natl Acad Sci U S A 2016; 113:9816-21. [PMID: 27540116 PMCID: PMC5024586 DOI: 10.1073/pnas.1611189113] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Clathrin, a cytosolic protein composed of heavy and light chain subunits, assembles into a vesicle coat, controlling receptor-mediated endocytosis. To establish clathrin light chain (CLC) function in vivo, we engineered mice lacking CLCa, the major CLC isoform in B lymphocytes, generating animals with CLC-deficient B cells. In CLCa-null mice, the germinal centers have fewer B cells, and they are enriched for IgA-producing cells. This enhanced switch to IgA production in the absence of CLCa was attributable to increased transforming growth factor β receptor 2 (TGFβR2) signaling resulting from defective endocytosis. Internalization of C-X-C chemokine receptor 4 (CXCR4), but not CXCR5, was affected in CLCa-null B cells, and CLC depletion from cell lines affected endocytosis of the δ-opioid receptor, but not the β2-adrenergic receptor, defining a role for CLCs in the uptake of a subset of signaling receptors. This instance of clathrin subunit deletion in vertebrates demonstrates that CLCs contribute to clathrin's role in vivo by influencing cargo selectivity, a function previously assigned exclusively to adaptor molecules.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- Cerebral Cortex/cytology
- Cerebral Cortex/immunology
- Clathrin Light Chains/genetics
- Clathrin Light Chains/immunology
- Endocytosis/immunology
- Gene Deletion
- Gene Expression Regulation
- Humans
- Immunoglobulin A/biosynthesis
- Immunoglobulin A/genetics
- Immunoglobulin Class Switching
- Liver/cytology
- Liver/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardium/cytology
- Myocardium/immunology
- Organ Specificity
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/immunology
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/immunology
- Receptors, CXCR4/genetics
- Receptors, CXCR4/immunology
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/immunology
- Receptors, Transforming Growth Factor beta/agonists
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/immunology
- Spleen/cytology
- Spleen/immunology
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Shuang Wu
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143; Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143; Department of Microbiology and Immunology, University of California, San Francisco, CA 94143; The G. W. Hooper Foundation, University of California, San Francisco, CA 94143
| | - Sophia R Majeed
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143; Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143; Department of Microbiology and Immunology, University of California, San Francisco, CA 94143; The G. W. Hooper Foundation, University of California, San Francisco, CA 94143
| | - Timothy M Evans
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143; Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143; Department of Microbiology and Immunology, University of California, San Francisco, CA 94143; The G. W. Hooper Foundation, University of California, San Francisco, CA 94143
| | - Marine D Camus
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143; Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143; Department of Microbiology and Immunology, University of California, San Francisco, CA 94143; The G. W. Hooper Foundation, University of California, San Francisco, CA 94143; Division of Biosciences, University College London, London WC1E 6BT, United Kingdom
| | - Nicole M L Wong
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143; Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143; Department of Microbiology and Immunology, University of California, San Francisco, CA 94143; The G. W. Hooper Foundation, University of California, San Francisco, CA 94143
| | - Yvette Schollmeier
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143; Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143; Department of Microbiology and Immunology, University of California, San Francisco, CA 94143; The G. W. Hooper Foundation, University of California, San Francisco, CA 94143
| | - Minjong Park
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143; Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143; Department of Microbiology and Immunology, University of California, San Francisco, CA 94143; The G. W. Hooper Foundation, University of California, San Francisco, CA 94143
| | - Jagan R Muppidi
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143
| | - Andrea Reboldi
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143
| | - Peter Parham
- Department of Structural Biology, Stanford University, Stanford, CA 94305; Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305
| | - Jason G Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143;
| | - Frances M Brodsky
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143; Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143; Department of Microbiology and Immunology, University of California, San Francisco, CA 94143; The G. W. Hooper Foundation, University of California, San Francisco, CA 94143; Division of Biosciences, University College London, London WC1E 6BT, United Kingdom;
| |
Collapse
|
56
|
Guzman E, Taylor G, Hope J, Herbert R, Cubillos-Zapata C, Charleston B. Transduction of skin-migrating dendritic cells by human adenovirus 5 occurs via an actin-dependent phagocytic pathway. J Gen Virol 2016; 97:2703-2718. [PMID: 27528389 PMCID: PMC5078831 DOI: 10.1099/jgv.0.000581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Dendritic cells (DC) are central to the initiation of immune responses, and various approaches have been used to target vaccines to DC in order to improve immunogenicity. Cannulation of lymphatic vessels allows for the collection of DC that migrate from the skin. These migrating DC are involved in antigen uptake and presentation following vaccination. Human replication-deficient adenovirus (AdV) 5 is a promising vaccine vector for delivery of recombinant antigens. Although the mechanism of AdV attachment and penetration has been extensively studied in permissive cell lines, few studies have addressed the interaction of AdV with DC. In this study, we investigated the interaction of bovine skin-migrating DC and replication-deficient AdV-based vaccine vectors. We found that, despite lack of expression of Coxsackie B–Adenovirus Receptor and other known adenovirus receptors, AdV readily enters skin-draining DC via an actin-dependent endocytosis. Virus exit from endosomes was pH independent, and neutralizing antibodies did not prevent virus entry but did prevent virus translocation to the nucleus. We also show that combining adenovirus with adjuvant increases the absolute number of intracellular virus particles per DC but not the number of DC containing intracellular virus. This results in increased trans-gene expression and antigen presentation. We propose that, in the absence of Coxsackie B–Adenovirus Receptor and other known receptors, AdV5-based vectors enter skin-migrating DC using actin-dependent endocytosis which occurs in skin-migrating DC, and its relevance to vaccination strategies and vaccine vector targeting is discussed.
Collapse
Affiliation(s)
- Efrain Guzman
- The Pirbright Institute, Ash Road, Woking, Surrey GU240NF, UK
| | | | - Jayne Hope
- The Roslin Institute University of Edinburgh, Easter Bush, Midlothian EH259RG, UK
| | - Rebecca Herbert
- The Pirbright Institute, Ash Road, Woking, Surrey GU240NF, UK
| | | | | |
Collapse
|
57
|
Wu CJ, Lu CH, Chen LC, Nguyen DT, Huang YS, Lin HH, Lin CY, Kuo ML. CD4 down regulation and raft dissociation by the non-depleting YTS177 antibody hinder murine T helper cell activities. Biochem Biophys Res Commun 2016; 473:973-979. [PMID: 27045081 DOI: 10.1016/j.bbrc.2016.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 04/01/2016] [Indexed: 11/28/2022]
Abstract
Non-depleting YTS177 anti-CD4 monoclonal antibody (MoAb) has been reported to lead to antigen-specific immunotolerance in allograft transplantation and autoimmune diabetes, as well as possibly to inhibition of allergic inflammation in mice. However, the molecular mechanisms underlying hyporesponsive T cell responses induced by YTS177 MoAb remain elusive. Herein, we demonstrate that the YTS177 MoAb increases the levels of anergy factors p27(kip1) and Cbl-b, inhibits IL-2 production, and impairs calcium mobilization in activated T cells in vitro. YTS177 MoAb suppresses OVA-driven proliferation of DO11.10 CD4(+) T cells in vivo as well. Mechanistically, YTS177 MoAb induces tolerance by causing CD4 down-regulation through clathrin-dependent and raft dissociation. The results obtained in this study lead us to propose novel protective or curative approaches to CD4 T cell-mediated diseases.
Collapse
Affiliation(s)
- Cheng-Jang Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan; Division of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Chun-Hao Lu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Li-Chen Chen
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Duc T Nguyen
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yi-Shu Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Hsi-Hsien Lin
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan; Department of Anatomic Pathology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Chun-Yen Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan; Department of Hepatogastroenterology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Ming-Ling Kuo
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan; Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan.
| |
Collapse
|
58
|
Huang SS, Chen CL, Huang FW, Hou WH, Huang JS. DMSO Enhances TGF-β Activity by Recruiting the Type II TGF-β Receptor From Intracellular Vesicles to the Plasma Membrane. J Cell Biochem 2016; 117:1568-79. [PMID: 26587792 DOI: 10.1002/jcb.25448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/18/2015] [Indexed: 01/03/2023]
Abstract
Dimethyl sulfoxide (DMSO) is used to treat many diseases/symptoms. The molecular basis of the pharmacological actions of DMSO has been unclear. We hypothesized that DMSO exerts some of these actions by enhancing TGF-β activity. Here we show that DMSO enhances TGF-β activity by ∼3-4-fold in Mv1Lu and NMuMG cells expressing Smad-dependent luciferase reporters. In Mv1Lu cells, DMSO enhances TGF-β-stimulated expression of P-Smad2 and PAI-1. It increases cell-surface expression of TGF-β receptors (TβR-I and/or TβR-II) by ∼3-4-fold without altering their cellular levels as determined by (125) I-labeled TGF-β-cross-linking/Western blot analysis, suggesting the presence of large intracellular pools in these cells. Sucrose density gradient ultracentrifugation/Western blot analysis reveals that DMSO induces recruitment of TβR-II (but not TβR-I) from its intracellular pool to plasma-membrane microdomains. It induces more recruitment of TβR-II to non-lipid raft microdomains than to lipid rafts/caveolae. Mv1Lu cells transiently transfected with TβR-II-HA plasmid were treated with DMSO and analyzed by indirect immunofluoresence staining using anti-HA antibody. In these cells, TβR-II-HA is present as a vesicle-like network in the cytoplasm as well as in the plasma membrane. DMSO causes depletion of TβR-II-HA-containing vesicles from the cytoplasm and co-localization of TβR-II-HA and cveolin-1 at the plasma membrane. These results suggest that DMSO, a fusogenic substance, enhances TGF-β activity presumably by inducing fusion of cytoplasmic vesicles (containing TβR-II) and the plasma membrane, resulting in increased localization of TβR-II to non-lipid raft microdomains where canonical signaling occurs. Fusogenic activity of DMSO may play a pivotal role in its pharmacological actions involving membrane proteins with large cytoplasmic pools. J. Cell. Biochem. 117: 1568-1579, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-sen University and Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 804, Taiwan
| | - Franklin W Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston and Harvard Medical School, Boston, Massachusetts, 02115
| | - Wei-Hsien Hou
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, 1100 S. Grand Boulevard, St. Louis, Missouri, 63104
| | - Jung San Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, 1100 S. Grand Boulevard, St. Louis, Missouri, 63104
| |
Collapse
|
59
|
Amsalem AR, Marom B, Shapira KE, Hirschhorn T, Preisler L, Paarmann P, Knaus P, Henis YI, Ehrlich M. Differential regulation of translation and endocytosis of alternatively spliced forms of the type II bone morphogenetic protein (BMP) receptor. Mol Biol Cell 2016; 27:716-30. [PMID: 26739752 PMCID: PMC4750929 DOI: 10.1091/mbc.e15-08-0547] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/24/2015] [Indexed: 12/22/2022] Open
Abstract
The cytoplasmic extension of the long-form isoform of BMPRII, unique among TGF-β superfamily receptors, is found to regulate the translation of BMPRII and its clathrin-mediated endocytosis. Both processes reduce its cell surface levels. The higher expression of BMPRII-SF at the plasma membrane results in enhanced activation of Smad signaling. The expression and function of transforming growth factor-β superfamily receptors are regulated by multiple molecular mechanisms. The type II BMP receptor (BMPRII) is expressed as two alternatively spliced forms, a long and a short form (BMPRII-LF and –SF, respectively), which differ by an ∼500 amino acid C-terminal extension, unique among TGF-β superfamily receptors. Whereas this extension was proposed to modulate BMPRII signaling output, its contribution to the regulation of receptor expression was not addressed. To map regulatory determinants of BMPRII expression, we compared synthesis, degradation, distribution, and endocytic trafficking of BMPRII isoforms and mutants. We identified translational regulation of BMPRII expression and the contribution of a 3’ terminal coding sequence to this process. BMPRII-LF and -SF differed also in their steady-state levels, kinetics of degradation, intracellular distribution, and internalization rates. A single dileucine signal in the C-terminal extension of BMPRII-LF accounted for its faster clathrin-mediated endocytosis relative to BMPRII-SF, accompanied by mildly faster degradation. Higher expression of BMPRII-SF at the plasma membrane resulted in enhanced activation of Smad signaling, stressing the potential importance of the multilayered regulation of BMPRII expression at the plasma membrane.
Collapse
Affiliation(s)
- Ayelet R Amsalem
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Barak Marom
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Keren E Shapira
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tal Hirschhorn
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Livia Preisler
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Pia Paarmann
- Institute for Chemistry and Biochemistry, Freie Univesitaet Berlin, 1495 Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Univesitaet Berlin, 1495 Berlin, Germany
| | - Yoav I Henis
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
60
|
Wang X, Schröder HC, Müller WEG. Polyphosphate as a metabolic fuel in Metazoa: A foundational breakthrough invention for biomedical applications. Biotechnol J 2016; 11:11-30. [PMID: 26356505 DOI: 10.1002/biot.201500168] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 07/24/2015] [Accepted: 08/19/2015] [Indexed: 12/17/2022]
Abstract
In animals, energy-rich molecules like ATP are generated in the intracellular compartment from metabolites, e.g. glucose, taken up by the cells. Recent results revealed that inorganic polyphosphates (polyP) can provide an extracellular system for energy transport and delivery. These polymers of multiple phosphate units, linked by high-energy phosphoanhydride bonds, use blood platelets as transport vehicles to reach their target cells. In this review it is outlined how polyP affects cell metabolism. It is discussed that polyP influences cell activity in a dual way: (i) as a metabolic fuel transferring metabolic energy through the extracellular space; and (ii) as a signaling molecule that amplifies energy/ATP production in mitochondria. Several metabolic pathways are triggered by polyP, among them biomineralization/hydroxyapatite formation onto bone cells. The accumulation of polyP in the platelets allows long-distance transport of the polymer in the extracellular space. The discovery of polyP as metabolic fuel and signaling molecule initiated the development of novel techniques for encapsulation of polyP into nanoparticles. They facilitate cellular uptake of the polymer by receptor-mediated endocytosis and allow the development of novel strategies for therapy of metabolic diseases associated with deviations in energy metabolism or mitochondrial dysfunctions.
Collapse
Affiliation(s)
- Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Rheinland-Pfalz, Germany.
| | - Heinz C Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Rheinland-Pfalz, Germany
| | - Werner E G Müller
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Rheinland-Pfalz, Germany.
| |
Collapse
|
61
|
Ehrlich M. Endocytosis and trafficking of BMP receptors: Regulatory mechanisms for fine-tuning the signaling response in different cellular contexts. Cytokine Growth Factor Rev 2015; 27:35-42. [PMID: 26776724 DOI: 10.1016/j.cytogfr.2015.12.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Signaling by bone morphogenetic protein (BMP) receptors is regulated at multiple levels in order to ensure proper interpretation of BMP stimuli in different cellular settings. As with other signaling receptors, regulation of the amount of exposed and signaling-competent BMP receptors at the plasma-membrane is predicted to be a key mechanism in governing their signaling output. Currently, the endocytosis of BMP receptors is thought to resemble that of the structurally related transforming growth factor-β (TGF-β) receptors, as BMP receptors are constitutively internalized (independently of ligand binding), with moderate kinetics, and mostly via clathrin-mediated endocytosis. Also similar to TGF-β receptors, BMP receptors are able to signal from the plasma membrane, while internalization to endosomes may have a signal modulating effect. When at the plasma membrane, BMP receptors localize to different membrane domains including cholesterol rich domains and caveolae, suggesting a complex interplay between membrane distribution and internalization. An additional layer of complexity stems from the putative regulatory influence on the signaling and trafficking of BMP receptors exerted by ligand traps and/or co-receptors. Furthermore, the trafficking and signaling of BMP receptors are subject to alterations in cellular context. For example, genetic diseases involving changes in the expression of auxiliary factors of endocytic pathways hamper retrograde BMP signals in neurons, and perturb the regulation of synapse formation. This review summarizes current understanding of the trafficking of BMP receptors and discusses the role of trafficking in regulation of BMP signals.
Collapse
Affiliation(s)
- Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
62
|
Wang C, Wang Y, Hu M, Chai Z, Wu Q, Huang R, Han W, Zhang CX, Zhou Z. Synaptotagmin-11 inhibits clathrin-mediated and bulk endocytosis. EMBO Rep 2015; 17:47-63. [PMID: 26589353 DOI: 10.15252/embr.201540689] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/21/2015] [Indexed: 11/09/2022] Open
Abstract
Precise and efficient endocytosis is essential for vesicle recycling during a sustained neurotransmission. The regulation of endocytosis has been extensively studied, but inhibitors have rarely been found. Here, we show that synaptotagmin-11 (Syt11), a non-Ca(2+)-binding Syt implicated in schizophrenia and Parkinson's disease, inhibits clathrin-mediated endocytosis (CME) and bulk endocytosis in dorsal root ganglion neurons. The frequency of both types of endocytic event increases in Syt11 knockdown neurons, while the sizes of endocytosed vesicles and the kinetics of individual bulk endocytotic events remain unaffected. Specifically, clathrin-coated pits and bulk endocytosis-like structures increase on the plasma membrane in Syt11-knockdown neurons. Structural-functional analysis reveals distinct domain requirements for Syt11 function in CME and bulk endocytosis. Importantly, Syt11 also inhibits endocytosis in hippocampal neurons, implying a general role of Syt11 in neurons. Taken together, we propose that Syt11 functions to ensure precision in vesicle retrieval, mainly by limiting the sites of membrane invagination at the early stage of endocytosis.
Collapse
Affiliation(s)
- Changhe Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China College of Life Sciences, Forestry and Agriculture, Qiqihar University, Qiqihar, China
| | - Yeshi Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Meiqin Hu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zuying Chai
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Qihui Wu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Rong Huang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Weiping Han
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, Agency for Science, Technology, and Research, Singapore City, Singapore Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Claire Xi Zhang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
63
|
Popli P, Sirohi VK, Manohar M, Shukla V, Kaushal JB, Gupta K, Dwivedi A. Regulation of cyclooxygenase-2 expression in rat oviductal epithelial cells: Evidence for involvement of GPR30/Src kinase-mediated EGFR signaling. J Steroid Biochem Mol Biol 2015; 154:130-41. [PMID: 26241029 DOI: 10.1016/j.jsbmb.2015.07.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/13/2015] [Accepted: 07/29/2015] [Indexed: 11/26/2022]
Abstract
The oviduct plays a crucial role in female reproduction by regulating gamete transport, providing a specific microenvironment for fertilization and early embryonic development. Cyclooxygenase (COX)-derived prostaglandins play essential role in carrying out these oviduct-specific functions. Estrogen upregulates COX-2 expression in rat oviduct; however, the mechanisms responsible for regulation of COX-2 expression in rat oviductal epithelial cells (OECs) remain unclear. In the present study, we proposed that estrogen induces COX-2 expression via G-protein coupled receptor i.e., GPR30 in OECs. To investigate this hypothesis, we examined the effects of E2-BSA, ICI 182,780, GPR30 agonist and GPR30 antagonist on COX-2 expression and explored potential signaling pathway leading to COX-2 expression. Co-localization experiments revealed GPR30 to be primarily located in the peri-nuclear space, which was also the site of E2-BSA-fluorescein isothiocyanate (E2-BSA-FITC) binding. The E2-BSA induced-COX-2 and prostaglandin release were subjected to regulation by both EGFR and PI3K signaling as inhibitors of c-Src kinase (PP2), EGFR (EGFR inhibitor) and PI-3 kinase (LY294002) attenuated E2-BSA mediated effect. These results suggest that EGFR transactivation leading to activation of PI-3K/Akt pathway participates in COX-2 expression in rat OECs. Interestingly, E2-BSA induced COX-2 expression and subsequent prostaglandin release were abolished by NF-κB inhibitor. In addition, E2-BSA induced the nuclear translocation of p65-NF-κB and up-regulated the NF-κB promoter activity in rat OECs. Taken together, results demonstrated that E2-BSA induced the COX-2 expression and consequent PGE2 and PGF2α release in rat OECs. These effects are mediated through GPR30-derived EGFR transactivation and PI-3K/Akt cascade leading to NF-κB activation.
Collapse
Affiliation(s)
- Pooja Popli
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Vijay Kumar Sirohi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Murli Manohar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Vinay Shukla
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Jyoti Bala Kaushal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Kanchan Gupta
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Anila Dwivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India.
| |
Collapse
|
64
|
Huang SS, Chen CL, Huang FW, Johnson FE, Huang JS. Ethanol Enhances TGF-β Activity by Recruiting TGF-β Receptors From Intracellular Vesicles/Lipid Rafts/Caveolae to Non-Lipid Raft Microdomains. J Cell Biochem 2015; 117:860-71. [PMID: 26419316 DOI: 10.1002/jcb.25389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022]
Abstract
Regular consumption of moderate amounts of ethanol has important health benefits on atherosclerotic cardiovascular disease (ASCVD). Overindulgence can cause many diseases, particularly alcoholic liver disease (ALD). The mechanisms by which ethanol causes both beneficial and harmful effects on human health are poorly understood. Here we demonstrate that ethanol enhances TGF-β-stimulated luciferase activity with a maximum of 0.5-1% (v/v) in Mv1Lu cells stably expressing a luciferase reporter gene containing Smad2-dependent elements. In Mv1Lu cells, 0.5% ethanol increases the level of P-Smad2, a canonical TGF-β signaling sensor, by ∼ 2-3-fold. Ethanol (0.5%) increases cell-surface expression of the type II TGF-β receptor (TβR-II) by ∼ 2-3-fold from its intracellular pool, as determined by I(125) -TGF-β-cross-linking/Western blot analysis. Sucrose density gradient ultracentrifugation and indirect immunofluorescence staining analyses reveal that ethanol (0.5% and 1%) also displaces cell-surface TβR-I and TβR-II from lipid rafts/caveolae and facilitates translocation of these receptors to non-lipid raft microdomains where canonical signaling occurs. These results suggest that ethanol enhances canonical TGF-β signaling by increasing non-lipid raft microdomain localization of the TGF-β receptors. Since TGF-β plays a protective role in ASCVD but can also cause ALD, the TGF-β enhancer activity of ethanol at low and high doses appears to be responsible for both beneficial and harmful effects. Ethanol also disrupts the location of lipid raft/caveolae of other membrane proteins (e.g., neurotransmitter, growth factor/cytokine, and G protein-coupled receptors) which utilize lipid rafts/caveolae as signaling platforms. Displacement of these membrane proteins induced by ethanol may result in a variety of pathologies in nerve, heart and other tissues.
Collapse
Affiliation(s)
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung, 804, Taiwan.,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 804, Taiwan
| | - Franklin W Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, 02115.,Harvard Medical School, Boston, Massachusetts, 02115
| | - Frank E Johnson
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri, 63104
| | - Jung San Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, St. Louis, Missouri, 63104
| |
Collapse
|
65
|
Le Guével X, Perez Perrino M, Fernández TD, Palomares F, Torres MJ, Blanca M, Rojo J, Mayorga C. Multivalent Glycosylation of Fluorescent Gold Nanoclusters Promotes Increased Human Dendritic Cell Targeting via Multiple Endocytic Pathways. ACS APPLIED MATERIALS & INTERFACES 2015; 7:20945-56. [PMID: 26329370 DOI: 10.1021/acsami.5b06541] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
We report the synthesis and characterization of gold nanoclusters (Au NCs) stabilized by a mixture of zwitterionic and multivalent mannose ligands. Characterization of this carbohydrated nanosystem confirms its small size (∼2 nm), intense red-NIR fluorescence, relatively high affinity to lectin (ConA), and stability in physiological media. Cell studies performed using human-monocyte-derived dendritic cells (DCs) show that Au NC uptake efficiency is greatly enhanced by the presence of surface carbohydrate (>250% compared to noncarbohydrated Au NCs), allowing their detection in cells by fluorescence following incubation with concentrations as low as 1 μg mL(-1). Investigation using electron microscopy and pharmacological inhibitors indicates that Au NC uptake is mediated by multiple endocytic pathways involving the engulfment of Au NCs into endosomes and partial transport to lysosomes. Results show that clathrin- and F-actin-dependent pathways play major roles in Au NC uptake by DCs, regardless of whether or not they are coated with carbohydrates. In contrast, a specific C-lectin inhibitor induces a 60% decrease in DC particle uptake only for the carbohydrate-coated Au NCs. This study demonstrates that the combination of ultrasmall gold NCs and functionalization with multivalent mannose ligands results in greatly enhanced human DC targeting, presumably due to increased diffusion and target cell binding, respectively.
Collapse
Affiliation(s)
- Xavier Le Guével
- Therapeutic Nanosystems, The Andalusian Centre for Nanomedicine and Biotechnology (BIONAND) , 29590 Málaga, Spain
| | - Monica Perez Perrino
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC-University of Seville , 41092 Seville, Spain
| | | | | | | | | | - Javier Rojo
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC-University of Seville , 41092 Seville, Spain
| | | |
Collapse
|
66
|
Müller WEG, Tolba E, Schröder HC, Wang X. Polyphosphate: A Morphogenetically Active Implant Material Serving as Metabolic Fuel for Bone Regeneration. Macromol Biosci 2015; 15:1182-1197. [DOI: 10.1002/mabi.201500100] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Werner E. G. Müller
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry; University Medical Center of the Johannes Gutenberg University; Duesbergweg 6; D-55128 Mainz Germany
| | - Emad Tolba
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry; University Medical Center of the Johannes Gutenberg University; Duesbergweg 6; D-55128 Mainz Germany
- Biomaterials Department; Inorganic Chemical Industries Division; National Research Center; Doki Cairo; 11884 Egypt
| | - Heinz C. Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry; University Medical Center of the Johannes Gutenberg University; Duesbergweg 6; D-55128 Mainz Germany
| | - Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry; University Medical Center of the Johannes Gutenberg University; Duesbergweg 6; D-55128 Mainz Germany
| |
Collapse
|
67
|
Autophagy Activated by Bluetongue Virus Infection Plays a Positive Role in Its Replication. Viruses 2015; 7:4657-75. [PMID: 26287233 PMCID: PMC4576199 DOI: 10.3390/v7082838] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/27/2015] [Accepted: 07/30/2015] [Indexed: 01/19/2023] Open
Abstract
Bluetongue virus (BTV) is an important pathogen of wild and domestic ruminants. Despite extensive study in recent decades, the interplay between BTV and host cells is not clearly understood. Autophagy as a cellular adaptive response plays a part in many viral infections. In our study, we found that BTV1 infection triggers the complete autophagic process in host cells, as demonstrated by the appearance of obvious double-membrane autophagosome-like vesicles, GFP-LC3 dots accumulation, the conversion of LC3-I to LC3-II and increased levels of autophagic flux in BSR cells (baby hamster kidney cell clones) and primary lamb lingual epithelial cells upon BTV1 infection. Moreover, the results of a UV-inactivated BTV1 infection assay suggested that the induction of autophagy was dependent on BTV1 replication. Therefore, we investigated the role of autophagy in BTV1 replication. The inhibition of autophagy by pharmacological inhibitors (3-MA, CQ) and RNA interference (siBeclin1) significantly decreased viral protein synthesis and virus yields. In contrast, treating BSR cells with rapamycin, an inducer of autophagy, promoted viral protein expression and the production of infectious BTV1. These findings lead us to conclude that autophagy is activated by BTV1 and contributes to its replication, and provide novel insights into BTV-host interactions.
Collapse
|
68
|
Jane-wit D, Surovtseva YV, Qin L, Li G, Liu R, Clark P, Manes TD, Wang C, Kashgarian M, Kirkiles-Smith NC, Tellides G, Pober JS. Complement membrane attack complexes activate noncanonical NF-κB by forming an Akt+ NIK+ signalosome on Rab5+ endosomes. Proc Natl Acad Sci U S A 2015; 112:9686-91. [PMID: 26195760 PMCID: PMC4534258 DOI: 10.1073/pnas.1503535112] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Complement membrane attack complexes (MACs) promote inflammatory functions in endothelial cells (ECs) by stabilizing NF-κB-inducing kinase (NIK) and activating noncanonical NF-κB signaling. Here we report a novel endosome-based signaling complex induced by MACs to stabilize NIK. We found that, in contrast to cytokine-mediated activation, NIK stabilization by MACs did not involve cIAP2 or TRAF3. Informed by a genome-wide siRNA screen, instead this response required internalization of MACs in a clathrin-, AP2-, and dynamin-dependent manner into Rab5(+)endosomes, which recruited activated Akt, stabilized NIK, and led to phosphorylation of IκB kinase (IKK)-α. Active Rab5 was required for recruitment of activated Akt to MAC(+) endosomes, but not for MAC internalization or for Akt activation. Consistent with these in vitro observations, MAC internalization occurred in human coronary ECs in vivo and was similarly required for NIK stabilization and EC activation. We conclude that MACs activate noncanonical NF-κB by forming a novel Akt(+)NIK(+) signalosome on Rab5(+) endosomes.
Collapse
Affiliation(s)
- Dan Jane-wit
- Division of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Yulia V Surovtseva
- Yale Center for Molecular Discovery, Yale University, New Haven, CT 06516
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520
| | - Guangxin Li
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520
| | - Rebecca Liu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Pamela Clark
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Thomas D Manes
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Chen Wang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Michael Kashgarian
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520
| | | | - George Tellides
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520; Department of Pathology, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
69
|
Phosphatidic Acid Increases Epidermal Growth Factor Receptor Expression by Stabilizing mRNA Decay and by Inhibiting Lysosomal and Proteasomal Degradation of the Internalized Receptor. Mol Cell Biol 2015; 35:3131-44. [PMID: 26124282 DOI: 10.1128/mcb.00286-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 06/22/2015] [Indexed: 12/18/2022] Open
Abstract
Overexpression of epidermal growth factor receptor (EGFR) is one of the frequent mechanisms implicated in cancer progression, and so is the overexpression of the enzyme phospholipase D (PLD) and its reaction product, phosphatidic acid (PA). However, an understanding of how these signaling molecules interact at the level of gene expression is lacking. Catalytically active PLD enhanced expression of EGFR in human breast cancer cells. Overexpression of the PLD2 isoform increased EGFR mRNA and protein expression. It also negated an EGFR downregulation mediated by small interfering RNA targeting EGFR (siEGFR). Several mechanisms contributed to the alteration in EGFR expression. First was the stabilization of EGFR transcripts as PLD2 delayed mRNA decay, which prolonged their half-lives. Second, RNase enzymatic activity was inhibited by PA. Third, protein stabilization also occurred, as indicated by PLD resistance to cycloheximide-induced EGFR protein degradation. Fourth, PA inhibited lysosomal and proteasomal degradation of internalized EGFR. PLD2 and EGFR colocalized at the cell membrane, and JAK3 phosphorylation at Tyr980/Tyr981 followed receptor endocytosis. Further, the presence of PLD2 increased stabilization of intracellular EGFR in large recycling vesicles at ∼15 min of EGF stimulation. Thus, PLD2-mediated production of PA contributed to the control of EGFR exposure to ligand through a multipronged transcriptional and posttranscriptional program during the out-of-control accumulation of EGFR signaling in cancer cells.
Collapse
|
70
|
Predescu S, Bardita C, Predescu D. New insights into the functions of intersectin-1s. Commun Integr Biol 2015; 8:e1034400. [PMID: 26479042 PMCID: PMC4594419 DOI: 10.1080/19420889.2015.1034400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 11/27/2022] Open
Abstract
Intersectin-1s (ITSN) is a ubiquitously expressed multifunctional protein known as a scaffold and regulator of the general endocytic machinery as well as a critical integrator of cellular signaling pathways. We showed recently that ITSN deficiency triggers a transforming growth factor β (TGFβ)/Alk5 signaling switch, from the canonical Smad 2/3 to the Erk1/2 MAPK pathway; moreover, endocytic impairment induced by ITSN deficiency enhances Alk5 ubiquitination and degradation and elicits TGFβ-paracrine effects mediated by circulating microparticles, leading to endothelial cell survival and increased proliferation. The studies expand our understanding of how ITSN facilitates cross-regulation of signaling pathways and provide insights into the involvement of ITSN deficiency in human disease.
Collapse
Affiliation(s)
- Sanda Predescu
- Departments of Pharmacology and Critical Care Medicine; Rush University Medical Center ; Chicago, IL USA
| | - Cristina Bardita
- Departments of Pharmacology and Critical Care Medicine; Rush University Medical Center ; Chicago, IL USA ; Department of Internal Medicine; Rush University Medical Center ; Chicago, IL USA
| | - Dan Predescu
- Departments of Pharmacology and Critical Care Medicine; Rush University Medical Center ; Chicago, IL USA
| |
Collapse
|
71
|
Saraf P, Li X, Wrischnik L, Jasti B. In Vitro and In Vivo Efficacy of Self-Assembling RGD Peptide Amphiphiles for Targeted Delivery of Paclitaxel. Pharm Res 2015; 32:3087-101. [PMID: 26063045 DOI: 10.1007/s11095-015-1689-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 03/26/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE The objective of this work was to compare the efficacy of self-assembling cyclic and linear RGD peptide amphiphiles as carriers for delivering paclitaxel to αvβ3 integrin overexpressing tumors. METHODS Linear (C18-ADA5-RGD) and cyclic (C18-ADA5-cRGDfK) peptide amphiphiles were synthesized and characterized for CMC, aggregation number and micelle stability using fluorescence spectroscopy methods. Size and morphology of micelles was studied using TEM. Fluorescence polarization and confocal microscopy assays were established to compare binding and internalization of micelles. The targeting efficacy was studied in A2058 cells using cytotoxicity assay as well as in vivo in melanoma xenograft mouse model. RESULTS The linear and cyclic RGD amphiphiles exhibited CMC of 25 and 8 μM, respectively, formed nano-sized spherical micelles and showed competitive binding to αvβ3 integrin protein. FITC-loaded RGD micelles rapidly internalized into A2058 melanoma cells. Paclitaxel-loaded RGD micelles exhibited higher cytotoxicity compared with free drug in A2058 cells in vitro as well as in vivo. CONCLUSION Cyclic RGD micelles exhibited better targeting efficacy but were less effective compared to linear RGD micelles as drug delivery vehicle due to lower drug solubilization capacity and lesser kinetic stability. Results from the study proved the effectiveness of self-assembling low molecular weight RGD amphiphiles as carriers for targeted delivery of paclitaxel.
Collapse
Affiliation(s)
- Poonam Saraf
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, USA
| | | | | | | |
Collapse
|
72
|
Müller WEG, Tolba E, Schröder HC, Diehl-Seifert B, Wang X. Retinol encapsulated into amorphous Ca(2+) polyphosphate nanospheres acts synergistically in MC3T3-E1 cells. Eur J Pharm Biopharm 2015; 93:214-223. [PMID: 25900862 DOI: 10.1016/j.ejpb.2015.04.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/06/2015] [Accepted: 04/10/2015] [Indexed: 11/19/2022]
Abstract
Both the quality and quantity of collagen, the major structural component of the skin, decrease in aging skin. We succeeded to encapsulate retinol into amorphous inorganic polyphosphate (polyP) nanoparticles together with calcium ions ("aCa-polyP-NP"), under formation of amorphous Ca-polyP/retinol nanospheres ("retinol/aCa-polyP-NS"). The globular nanospheres are not cytotoxic, show an almost uniform size of ≈ 45 nm and have a retinol content of around 25%. Both components of those nanospheres, retinol and the aCa-polyP-NP, if administered together, caused a strong increase in proliferation of mouse calvaria MC3T3 cells. The expressions of collagen types I, II and III genes, but not the expression of collagen type V gene, were significantly enhanced if retinol is added together with aCa-polyP-NP. This synergistic effect was especially pronounced for the expression of the collagen type III gene. We propose that the synergistic effect of the retinol/aCa-polyP-NS on cell growth and collagen type III expression is induced via two routes, first through cellular uptake of the 45 nm nanospheres by clathrin-mediated endocytosis and second through extracellular disintegration of the nanospheres resulting in the release of retinol which is then taken up into the cells after binding to the retinal binding protein receptor.
Collapse
Affiliation(s)
- Werner E G Müller
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Emad Tolba
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany; Biomaterials Department, Inorganic Chemical Industries Division, National Research Center, Doki, 11884 Cairo, Egypt
| | - Heinz C Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany
| | | | - Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| |
Collapse
|
73
|
Müller WEG, Tolba E, Feng Q, Schröder HC, Markl JS, Kokkinopoulou M, Wang X. Amorphous Ca²⁺ polyphosphate nanoparticles regulate the ATP level in bone-like SaOS-2 cells. J Cell Sci 2015; 128:2202-2207. [PMID: 25908856 DOI: 10.1242/jcs.170605] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 04/16/2015] [Indexed: 01/08/2023] Open
Abstract
Polyphosphate (polyP) is a physiologically occurring polyanion that is synthesized especially in bone-forming osteoblast cells and blood platelets. We used amorphous polyP nanoparticles, complexed with Ca(2+), that have a globular size of ∼100 nm. Because polyP comprises inorganic orthophosphate units that are linked together through high-energy phosphoanhydride bonds, we questioned whether the observed morphogenetic effect, elicited by polyP, is correlated with the energy-generating machinery within the cells. We show that exposure of SaOS-2 osteoblast-like cells to polyP results in a strong accumulation of mitochondria and a parallel translocation of the polyP-degrading enzyme alkaline phosphatase to the cell surface. If SaOS-2 cells are activated by the mineralization activation cocktail (comprising β-glycerophosphate, ascorbic acid and dexamethasone) and additionally incubated with polyP, a tenfold intracellular increase of the ATP level occurs. Even more, in those cells, an intensified release of ATP into the extracellular space is also seen. We propose and conclude that polyP acts as metabolic fuel after the hydrolytic cleavage of the phosphoanhydride linkages, which contributes to hydroxyapatite formation on the plasma membranes of osteoblasts.
Collapse
Affiliation(s)
- Werner E G Müller
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany
| | - Emad Tolba
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany Biomaterials Department, Inorganic Chemical Industries Division, National Research Center, Doki 11884, Cairo, Egypt
| | - Qingling Feng
- Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Heinz C Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany
| | - Julia S Markl
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany
| | - Maria Kokkinopoulou
- Department of Zoology, Johannes Gutenberg-University, Johannes-von-Müller-Weg 6, D-55099 Mainz, Germany
| | - Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany
| |
Collapse
|
74
|
Schwab EH, Pohl TLM, Haraszti T, Schwaerzer GK, Hiepen C, Spatz JP, Knaus P, Cavalcanti-Adam EA. Nanoscale control of surface immobilized BMP-2: toward a quantitative assessment of BMP-mediated signaling events. NANO LETTERS 2015; 15:1526-1534. [PMID: 25668064 DOI: 10.1021/acs.nanolett.5b00315] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
In this work we determine the impact of surface density of immobilized BMP-2 on intracellular signal transduction. We use block copolymer micellar nanolithography to fabricate substrates with precisely spaced and tunable gold nanoparticle arrays carrying single BMP-2 molecules. We found that the immobilized growth factor triggers prolonged and elevated Smad signaling pathway activation compared to the same amount of soluble protein. This approach is suitable for achieving controlled and sustained local delivery of BMP-2 and other growth factors.
Collapse
Affiliation(s)
- Elisabeth H Schwab
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg , INF 253, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Bardita C, Predescu DN, Sha F, Patel M, Balaji G, Predescu SA. Endocytic deficiency induced by ITSN-1s knockdown alters the Smad2/3-Erk1/2 signaling balance downstream of Alk5. J Cell Sci 2015; 128:1528-41. [PMID: 25720380 PMCID: PMC4406123 DOI: 10.1242/jcs.163030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/17/2015] [Indexed: 12/11/2022] Open
Abstract
Recently, we demonstrated in cultured endothelial cells and in vivo that deficiency of an isoform of intersectin-1, ITSN-1s, impairs caveolae and clathrin-mediated endocytosis and functionally upregulates compensatory pathways and their morphological carriers (i.e. enlarged endocytic structures, membranous rings or tubules) that are normally underrepresented. We now show that these endocytic structures internalize the broadly expressed transforming growth factor β receptor I (TGFβ-RI or TGFBR1), also known as Alk5, leading to its ubiquitylation and degradation. Moreover, the apoptotic or activated vascular cells of the ITSN-1s-knockdown mice release Alk5-bearing microparticles to the systemic circulation. These interact with and transfer Alk5 to endocytosis-deficient endothelial cells, resulting in lung endothelial cell survival and phenotypic alteration towards proliferation through activation of Erk1 and Erk2 (also known as MAPK3 and MAPK1, respectively). We also show that non-productive assembly of the Alk5–Smad–SARA (Smad anchor for receptor activation, also known as ZFYVE9) signaling complex and preferential formation of the Alk5–mSos–Grb2 complex account for Erk1/2 activation downstream of Alk5 and proliferation of pulmonary endothelial cells. Taken together, our studies demonstrate a functional relationship between the intercellular transfer of Alk5 by microparticles and endothelial cell survival and proliferation, and define a novel molecular mechanism for TGFβ and Alk5-dependent Erk1/2MAPK signaling that is significant for proliferative signaling and abnormal growth.
Collapse
Affiliation(s)
- Cristina Bardita
- Department of Pharmacology, Rush University, Chicago, IL 60612, USA
| | - Dan N Predescu
- Department of Pharmacology, Rush University, Chicago, IL 60612, USA Pulmonary and Critical Care Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Fei Sha
- Department of Pharmacology, Rush University, Chicago, IL 60612, USA
| | - Monal Patel
- Department of Pharmacology, Rush University, Chicago, IL 60612, USA
| | - Ganesh Balaji
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sanda A Predescu
- Department of Pharmacology, Rush University, Chicago, IL 60612, USA Pulmonary and Critical Care Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
76
|
Hirschhorn T, di Clemente N, Amsalem AR, Pepinsky RB, Picard JY, Smorodinsky NI, Cate RL, Ehrlich M. Constitutive negative regulation in the processing of the anti-Müllerian hormone receptor II. J Cell Sci 2015; 128:1352-64. [PMID: 25663701 DOI: 10.1242/jcs.160143] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The levels and intracellular localization of wild-type transforming growth factor β superfamily (TGFβ-SF) receptors are tightly regulated by endocytic trafficking, shedding and degradation. In contrast, a main regulatory mechanism of mutation-bearing receptors involves their intracellular retention. Anti-Müllerian hormone receptor II (AMHRII, also known as AMHR2) is the type-II receptor for anti-Müllerian hormone (AMH), a TGFβ-SF ligand that mediates Müllerian duct regression in males. Here, we studied AMHRII processing and identified novel mechanisms of its constitutive negative regulation. Immunoblot analysis revealed that a significant portion of AMHRII was missing most of its extracellular domain (ECD) and, although glycosylated, was unfolded and retained in the endoplasmic reticulum. Exogenous expression of AMHRII, but not of type-II TGF-β receptor (TβRII, also known as TGFR2), resulted in its disulfide-bond-mediated homo-oligomerization and intracellular retention, and in a decrease in its AMH-binding capacity. At the plasma membrane, AMHRII differed from TβRII, forming high levels of non-covalent homomeric complexes, which exhibited a clustered distribution and restricted lateral mobility. This study identifies novel mechanisms of negative regulation of a type-II TGFβ-SF receptor through cleavage, intracellular retention and/or promiscuous disulfide-bond mediated homo-oligomerization.
Collapse
Affiliation(s)
- Tal Hirschhorn
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv, Israel 69978
| | - Nathalie di Clemente
- Université Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013 Paris, France CNRS, UMR 8251, Biologie Fonctionnelle et Adaptative, F-75013 Paris, France INSERM U1133, Physiologie de l'Axe Gonadotrope, F-75013 Paris, France
| | - Ayelet R Amsalem
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - R Blake Pepinsky
- Biogen-Idec, Inc., 14 Cambridge Center, Cambridge, MA 02142, USA
| | - Jean-Yves Picard
- INSERM U1133, Physiologie de l'Axe Gonadotrope, F-75013 Paris, France
| | - Nechama I Smorodinsky
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv, Israel 69978
| | - Richard L Cate
- INSERM U1133, Physiologie de l'Axe Gonadotrope, F-75013 Paris, France Boston University, 590 Commonwealth Avenue, Boston, MA 02215, USA
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv, Israel 69978
| |
Collapse
|
77
|
Nasrolahi Shirazi A, Tiwari RK, Oh D, Sullivan B, Kumar A, Beni Y, Parang K. Cyclic peptide-selenium nanoparticles as drug transporters. Mol Pharm 2014; 11:3631-3641. [PMID: 25184366 PMCID: PMC4186687 DOI: 10.1021/mp500364a] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/03/2014] [Accepted: 09/03/2014] [Indexed: 02/07/2023]
Abstract
A cyclic peptide composed of five tryptophan, four arginine, and one cysteine [W5R4C] was synthesized. The peptide was evaluated for generating cyclic peptide-capped selenium nanoparticles (CP-SeNPs) in situ. A physical mixing of the cyclic peptide with SeO3(-2) solution in water generated [W5R4C]-SeNPs via the combination of reducing and capping properties of amino acids in the peptide structure. Transmission electron microscopy (TEM) images showed that [W5R4C]-SeNPs were in the size range of 110-150 nm. Flow cytometry data revealed that a fluorescence-labeled phosphopeptide (F'-PEpYLGLD, where F' = fluorescein) and an anticancer drug (F'-dasatinib) exhibited approximately 25- and 9-times higher cellular uptake in the presence of [W5R4C]-SeNPs than those of F'-PEpYLGLD and dasatinib alone in human leukemia (CCRF-CEM) cells after 2 h of incubation, respectively. Confocal microscopy also exhibited higher cellular delivery of F'-PEpYLGLD and F'-dasatinib in the presence of [W5R4C]-SeNPs compared to the parent fluorescence-labeled drug alone in human ovarian adenocarcinoma (SK-OV-3) cells after 2 h of incubation at 37 °C. The antiproliferative activities of several anticancer drugs doxorubicin, gemcitabine, clofarabine, etoposide, camptothecin, irinotecan, epirubicin, fludarabine, dasatinib, and paclitaxel were improved in the presence of [W5R4C]-SeNPs (50 μM) by 38%, 49%, 36%, 36%, 31%, 30%, 30%, 28%, 24%, and 17%, respectively, after 48 h incubation in SK-OV-3 cells. The results indicate that CP-SeNPs can be potentially used as nanosized delivery tools for negatively charged biomolecules and anticancer drugs.
Collapse
Affiliation(s)
- Amir Nasrolahi Shirazi
- Chao
Family Comprehensice Cancer Center, School of Medicine, University of California, Irvine, Shanbrom Hall, 101 The City Drive South, Orange, California 92868, United States
- Chapman
University School of Pharmacy, Harry and Diane Rinker Health Science
Campus, Irvine, California 92618, United States
- Department
of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Rakesh K. Tiwari
- Chao
Family Comprehensice Cancer Center, School of Medicine, University of California, Irvine, Shanbrom Hall, 101 The City Drive South, Orange, California 92868, United States
- Chapman
University School of Pharmacy, Harry and Diane Rinker Health Science
Campus, Irvine, California 92618, United States
- Department
of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Donghoon Oh
- Department
of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Brian Sullivan
- Department
of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Anil Kumar
- Department
of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Yousef
A. Beni
- Department
of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Keykavous Parang
- Chao
Family Comprehensice Cancer Center, School of Medicine, University of California, Irvine, Shanbrom Hall, 101 The City Drive South, Orange, California 92868, United States
- Chapman
University School of Pharmacy, Harry and Diane Rinker Health Science
Campus, Irvine, California 92618, United States
- Department
of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, United States
| |
Collapse
|
78
|
Entry of a novel marine DNA virus, Singapore grouper iridovirus, into host cells occurs via clathrin-mediated endocytosis and macropinocytosis in a pH-dependent manner. J Virol 2014; 88:13047-63. [PMID: 25165116 DOI: 10.1128/jvi.01744-14] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Iridoviruses are nucleocytoplasmic DNA viruses which cause great economic losses in the aquaculture industry but also show significant threat to global biodiversity. However, a lack of host cells has resulted in poor progress in clarifying iridovirus behavior. We investigated the crucial events during virus entry using a combination of single-virus tracking and biochemical assays, based on the established virus-cell infection model for Singapore grouper iridovirus (SGIV). SGIV infection in host cells was strongly inhibited when cells were pretreated with drugs blocking clathrin-mediated endocytosis, including sucrose and chlorpromazine. Inhibition of key regulators of macropinocytosis, including Na(+)/H(+) exchanger, Rac1 GTPase, p21-activated kinase 1 (PAK1), protein kinase C (PKC), and myosin II, significantly reduced SGIV uptake. Cy5-labeled SGIV particles were observed to colocalize with clathrin and macropinosomes. In contrast, disruption of cellular cholesterol by methyl-β-cyclodextrin and nystatin had no effect on virus infection, suggesting that SGIV entered grouper cells via the clathrin-mediated endocytic pathway and macropinocytosis but not via caveola-dependent endocytosis. Furthermore, inhibitors of endosome acidification such as chloroquine and bafilomycin A1 blocked virus infection, indicating that SGIV entered cells in a pH-dependent manner. In addition, SGIV particles were observed to be transported along both microtubules and actin filaments, and intracellular SGIV motility was remarkably impaired by depolymerization of microtubules or actin filaments. The results of this study for the first time demonstrate that not only the clathrin-dependent pathway but also macropinocytosis are involved in fish DNA enveloped virus entry, thus providing a convenient tactic for exploring the life cycle of DNA viruses. IMPORTANCE Virus entry into host cells is critically important for initiating infections and is usually recognized as an ideal target for the design of antiviral strategies. Iridoviruses are large DNA viruses which cause serious threats to ecological diversity and the aquaculture industry worldwide. However, the current understanding of iridovirus entry is limited and controversial. Singapore grouper iridovirus (SGIV) is a novel marine fish DNA virus which belongs to genus Ranavirus, family Iridoviridae. Here, using single-virus tracking technology in combination with biochemical assays, we investigated the crucial events during SGIV entry and demonstrated that SGIV entered grouper cells via the clathrin-mediated endocytic pathway in a pH-dependent manner but not via caveola-dependent endocytosis. Furthermore, we propose for the first time that macropinocytosis is involved in iridovirus entry. Together, this work not only contributes greatly to understating iridovirus pathogenesis but also provides an ideal model for exploring the behavior of DNA viruses in living cells.
Collapse
|
79
|
Hovater MB, Ying WZ, Agarwal A, Sanders PW. Nitric oxide and carbon monoxide antagonize TGF-β through ligand-independent internalization of TβR1/ALK5. Am J Physiol Renal Physiol 2014; 307:F727-35. [PMID: 25100282 DOI: 10.1152/ajprenal.00353.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Transforming growth factor (TGF)-β plays a central role in vascular homeostasis and in the pathology of vascular disease. There is a growing appreciation for the role of nitric oxide (NO) and carbon monoxide (CO) as highly diffusible, bioactive signaling molecules in the vasculature. We hypothesized that both NO and CO increase endocytosis of TGF-β receptor type 1 (TβR1) in vascular smooth muscle cells (VSMCs) through activation of dynamin-2, shielding cells from the effects of circulating TGF-β. In this study, primary cultures of VSMCs from Sprague-Dawley rats were treated with NO-releasing molecule 3 (a NO chemical donor), CO-releasing molecule 2 (a CO chemical donor), or control. NO and CO stimulated dynamin-2 activation in VSMCs. NO and CO promoted time- and dose-dependent endocytosis of TβR1. By decreasing TβR1 surface expression through this dynamin-2-dependent process, NO and CO diminished the effects of TGF-β on VSMCs. These findings help explain an important mechanism by which NO and CO signal in the vasculature by decreasing surface expression of TβR1 and the cellular response to TGF-β.
Collapse
Affiliation(s)
- Michael B Hovater
- Department of Medicine University of Alabama at Birmingham, Birmingham, Alabama
| | - Wei-Zhong Ying
- Department of Medicine University of Alabama at Birmingham, Birmingham, Alabama
| | - Anupam Agarwal
- Division of Nephrology, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Medicine University of Alabama at Birmingham, Birmingham, Alabama; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama; Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Veterans Affairs Medical Center, Birmingham, Alabama
| | - Paul W Sanders
- Division of Nephrology, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Medicine University of Alabama at Birmingham, Birmingham, Alabama; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama; Department of Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
80
|
Choi H, Nguyen HN, Lamb FS. Inhibition of endocytosis exacerbates TNF-α-induced endothelial dysfunction via enhanced JNK and p38 activation. Am J Physiol Heart Circ Physiol 2014; 306:H1154-63. [DOI: 10.1152/ajpheart.00885.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tumor necrosis factor-α (TNF-α) is a pro-inflammatory cytokine that causes endothelial dysfunction. Endocytosis of TNF-α receptors (TNFR) precedes endosomal reactive oxygen species (ROS) production, which is required for NF-κB activation in vascular smooth muscle cells. It is unknown how endocytosis of TNFRs impacts signaling in endothelial cells. We hypothesized that TNF-α-induced endothelial dysfunction is induced by both endosomal and cell surface events, including NF-κB and mitogen-activated protein kinases (MAPKs) activation, and endocytosis of the TNFR modifies signaling. Mesenteric artery segments from C57BL/6 mice were treated with TNF-α (10 ng/ml) for 22 h in tissue culture, with or without signaling inhibitors (dynasore for endocytosis, SP600125 for JNK, SB203580 for p38, U0126 for ERK), and vascular function was assessed. Endothelium-dependent relaxation to acetylcholine (ACh) was impaired by TNF-α, and dynasore exacerbated this, whereas JNK or p38 inhibition prevented these effects. In cultured endothelial cells from murine mesenteric arteries, dynasore potentiated JNK and p38 but not ERK phosphorylation and promoted cell death. NF-κB activation by TNF-α was decreased by dynasore. JNK inhibition dramatically increased both the magnitude and duration of TNF-α-induced NF-κB activation and potentiated intercellular adhesion molecule-1 (ICAM-1) activation. Dynasore still inhibited NF-κB activation in the presence of SP600125. Thus TNF-α-induced endothelial dysfunction is both JNK and p38 dependent. Endocytosis modulates the balance of NF-κB and MAPK signaling, and inhibition of NF-κB activation by JNK limits this pro-proliferative signal, which may contribute to endothelial cell death in response to TNF-α.
Collapse
Affiliation(s)
- Hyehun Choi
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hong N. Nguyen
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Fred S. Lamb
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
81
|
Shapira KE, Hirschhorn T, Barzilay L, Smorodinsky NI, Henis YI, Ehrlich M. Dab2 inhibits the cholesterol-dependent activation of JNK by TGF-β. Mol Biol Cell 2014; 25:1620-8. [PMID: 24648493 PMCID: PMC4019493 DOI: 10.1091/mbc.e13-09-0537] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
TGF-β signals through Smad-dependent and non-Smad pathways, depending on cell context. In ovarian cancer cells, the clathrin adaptor Dab2 enhances internalization of the type I TGF-β receptor, restricts its lateral mobility, and inhibits TGF-β–mediated, cholesterol-dependent JNK activation. Transforming growth factor-β (TGF-β) ligands activate Smad-mediated and noncanonical signaling pathways in a cell context–dependent manner. Localization of signaling receptors to distinct membrane domains is a potential source of signaling output diversity. The tumor suppressor/endocytic adaptor protein disabled-2 (Dab2) was proposed as a modulator of TGF-β signaling. However, the molecular mechanism(s) involved in the regulation of TGF-β signaling by Dab2 were not known. Here we investigate these issues by combining biophysical studies of the lateral mobility and endocytosis of the type I TGF-β receptor (TβRI) with TGF-β phosphoprotein signaling assays. Our findings demonstrate that Dab2 interacts with TβRI to restrict its lateral diffusion at the plasma membrane and enhance its clathrin-mediated endocytosis. Small interfering RNA–mediated knockdown of Dab2 or Dab2 overexpression shows that Dab2 negatively regulates TGF-β–induced c-Jun N-terminal kinase (JNK) activation, whereas activation of the Smad pathway is unaffected. Moreover, activation of JNK by TGF-β in the absence of Dab2 is disrupted by cholesterol depletion. These data support a model in which Dab2 regulates the domain localization of TβRI in the membrane, balancing TGF-β signaling via the Smad and JNK pathways.
Collapse
Affiliation(s)
- Keren E Shapira
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tal Hirschhorn
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lior Barzilay
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nechama I Smorodinsky
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoav I Henis
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
82
|
BMP signaling requires retromer-dependent recycling of the type I receptor. Proc Natl Acad Sci U S A 2014; 111:2578-83. [PMID: 24550286 DOI: 10.1073/pnas.1319947111] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The transforming growth factor β (TGFβ) superfamily of signaling pathways, including the bone morphogenetic protein (BMP) subfamily of ligands and receptors, controls a myriad of developmental processes across metazoan biology. Transport of the receptors from the plasma membrane to endosomes has been proposed to promote TGFβ signal transduction and shape BMP-signaling gradients throughout development. However, how postendocytic trafficking of BMP receptors contributes to the regulation of signal transduction has remained enigmatic. Here we report that the intracellular domain of Caenorhabditis elegans BMP type I receptor SMA-6 (small-6) binds to the retromer complex, and in retromer mutants, SMA-6 is degraded because of its missorting to lysosomes. Surprisingly, we find that the type II BMP receptor, DAF-4 (dauer formation-defective-4), is retromer-independent and recycles via a distinct pathway mediated by ARF-6 (ADP-ribosylation factor-6). Importantly, we find that loss of retromer blocks BMP signaling in multiple tissues. Taken together, our results indicate a mechanism that separates the type I and type II receptors during receptor recycling, potentially terminating signaling while preserving both receptors for further rounds of activation.
Collapse
|
83
|
Lum M, Attridge SR, Morona R. Impact of dynasore an inhibitor of dynamin II on Shigella flexneri infection. PLoS One 2013; 8:e84975. [PMID: 24367704 PMCID: PMC3868620 DOI: 10.1371/journal.pone.0084975] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 11/27/2013] [Indexed: 11/19/2022] Open
Abstract
Shigella flexneri remains a significant human pathogen due to high morbidity among children < 5 years in developing countries. One of the key features of Shigella infection is the ability of the bacterium to initiate actin tail polymerisation to disseminate into neighbouring cells. Dynamin II is associated with the old pole of the bacteria that is associated with F-actin tail formation. Dynamin II inhibition with dynasore as well as siRNA knockdown significantly reduced Shigella cell to cell spreading in vitro. The ocular mouse Sereny model was used to determine if dynasore could delay the progression of Shigella infection in vivo. While dynasore did not reduce ocular inflammation, it did provide significant protection against weight loss. Therefore dynasore's effects in vivo are unlikely to be related to the inhibition of cell spreading observed in vitro. We found that dynasore decreased S. flexneri-induced HeLa cell death in vitro which may explain the protective effect observed in vivo. These results suggest the administration of dynasore or a similar compound during Shigella infection could be a potential intervention strategy to alleviate disease symptoms.
Collapse
Affiliation(s)
- Mabel Lum
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Stephen R. Attridge
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Renato Morona
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
84
|
Organista-Juárez D, Carretero-Ortega J, Vicente-Fermín O, Vázquez-Victorio G, Sosa-Garrocho M, Vázquez-Prado J, Macías-Silva M, Reyes-Cruz G. Calcium-sensing receptor inhibits TGF-β-signaling by decreasing Smad2 phosphorylation. IUBMB Life 2013; 65:1035-42. [DOI: 10.1002/iub.1232] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/12/2013] [Indexed: 12/13/2022]
Affiliation(s)
- Diana Organista-Juárez
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Jorge Carretero-Ortega
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Onasis Vicente-Fermín
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Genaro Vázquez-Victorio
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Marcela Sosa-Garrocho
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - José Vázquez-Prado
- Department of Pharmacology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Marina Macías-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Guadalupe Reyes-Cruz
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| |
Collapse
|
85
|
Ashoor R, Yafawi R, Jessen B, Lu S. The contribution of lysosomotropism to autophagy perturbation. PLoS One 2013; 8:e82481. [PMID: 24278483 PMCID: PMC3838419 DOI: 10.1371/journal.pone.0082481] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/02/2013] [Indexed: 11/18/2022] Open
Abstract
Autophagy refers to the catabolic process in eukaryotic cells that delivers cytoplasmic material to lysosomes for degradation. This highly conserved process is involved in the clearance of long-lived proteins and damaged organelles. Consequently, autophagy is important in providing nutrients to maintain cellular function under starvation, maintaining cellular homeostasis, and promoting cell survival under certain conditions. Several pathways, including mTOR, have been shown to regulate autophagy. However, the impact of lysosomal function impairment on the autophagy process has not been fully explored. Basic lipophilic compounds can accumulate in lysosomes via pH partitioning leading to perturbation of lysosomal function. Our hypothesis is that these types of compounds can disturb the autophagy process. Eleven drugs previously shown to accumulate in lysosomes were selected and evaluated for their effects on cytotoxicity and autophagy using ATP depletion and LC3 assessment, respectively. All eleven drugs induced increased staining of endogenous LC3 and exogenous GFP-LC3, even at non toxic dose levels. In addition, an increase in the abundance of SQSTM1/p62 by all tested compounds denotes that the increase in LC3 is due to autophagy perturbation rather than enhancement. Furthermore, the gene expression profile resulting from in vitro treatment with these drugs revealed the suppression of plentiful long-lived proteins, including structural cytoskeletal and associated proteins, and extracellular matrix proteins. This finding indicates a retardation of protein turnover which further supports the notion of autophagy inhibition. Interestingly, upregulation of genes containing antioxidant response elements, e.g. glutathione S transferase and NAD(P)H dehydrogenase quinone 1 was observed, suggesting activation of Nrf2 transcription factor. These gene expression changes could be related to an increase in SQSTM1/p62 resulting from autophagy deficiency. In summary, our data indicate that lysosomal accumulation due to the basic lipophilic nature of xenobiotics could be a general mechanism contributing to the perturbation of the autophagy process.
Collapse
Affiliation(s)
- Roshan Ashoor
- Drug Safety Research and Development, Pfizer Inc., San Diego, California, United States of America
| | - Rolla Yafawi
- Drug Safety Research and Development, Pfizer Inc., San Diego, California, United States of America
| | - Bart Jessen
- Drug Safety Research and Development, Pfizer Inc., San Diego, California, United States of America
| | - Shuyan Lu
- Drug Safety Research and Development, Pfizer Inc., San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
86
|
Arezes J, Costa M, Vieira I, Dias V, Kong XL, Fernandes R, Vos M, Carlsson A, Rikers Y, Porto G, Rangel M, Hider RC, Pinto JP. Non-transferrin-bound iron (NTBI) uptake by T lymphocytes: evidence for the selective acquisition of oligomeric ferric citrate species. PLoS One 2013; 8:e79870. [PMID: 24278199 PMCID: PMC3836815 DOI: 10.1371/journal.pone.0079870] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 09/27/2013] [Indexed: 02/06/2023] Open
Abstract
Iron is an essential nutrient in several biological processes such as oxygen transport, DNA replication and erythropoiesis. Plasma iron normally circulates bound to transferrin. In iron overload disorders, however, iron concentrations exceed transferrin binding capacity and iron appears complexed with low molecular weight molecules, known as non-transferrin-bound iron (NTBI). NTBI is responsible for the toxicity associated with iron-overload pathologies but the mechanisms leading to NTBI uptake are not fully understood. Here we show for the first time that T lymphocytes are able to take up and accumulate NTBI in a manner that resembles that of hepatocytes. Moreover, we show that both hepatocytes and T lymphocytes take up the oligomeric Fe3Cit3 preferentially to other iron-citrate species, suggesting the existence of a selective NTBI carrier. These results provide a tool for the identification of the still elusive ferric-citrate cellular carrier and may also open a new pathway towards the design of more efficient iron chelators for the treatment of iron overload disorders.
Collapse
Affiliation(s)
- Joao Arezes
- Basic and Clinical Research on Iron Biology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- * E-mail:
| | - Monica Costa
- Basic and Clinical Research on Iron Biology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Ines Vieira
- Basic and Clinical Research on Iron Biology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Vera Dias
- Basic and Clinical Research on Iron Biology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Xiao L. Kong
- Pharmaceutical Sciences Research Division, King's College London, London, United Kingdom
| | - Rui Fernandes
- ATAF, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Matthijn Vos
- Europe NanoPort, FEI, Eindhoven, The Netherlands
| | | | - Yuri Rikers
- Europe NanoPort, FEI, Eindhoven, The Netherlands
| | - Graça Porto
- Basic and Clinical Research on Iron Biology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Clinical Hematology, CHP-HSA - Santo António General Hospital, Porto, Portugal
- Molecular Immunology and Pathology, ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria Rangel
- REQUIMTE, ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Robert C. Hider
- Pharmaceutical Sciences Research Division, King's College London, London, United Kingdom
| | - Jorge P. Pinto
- Basic and Clinical Research on Iron Biology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
87
|
Komatsu K, Shimada A, Shibata T, Wada S, Ideno H, Nakashima K, Amizuka N, Noda M, Nifuji A. Alendronate promotes bone formation by inhibiting protein prenylation in osteoblasts in rat tooth replantation model. J Endocrinol 2013; 219:145-58. [PMID: 24096963 DOI: 10.1530/joe-13-0040] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Bisphosphonates (BPs) are a major class of antiresorptive drug, and their molecular mechanisms of antiresorptive action have been extensively studied. Recent studies have suggested that BPs target bone-forming cells as well as bone-resorbing cells. We previously demonstrated that local application of a nitrogen-containing BP (N-BP), alendronate (ALN), for a short period of time increased bone tissue in a rat tooth replantation model. Here, we investigated cellular mechanisms of bone formation by ALN. Bone histomorphometry confirmed that bone formation was increased by local application of ALN. ALN increased proliferation of bone-forming cells residing on the bone surface, whereas it suppressed the number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts in vivo. Moreover, ALN treatment induced more alkaline phosphatase-positive and osteocalcin-positive cells on the bone surface than PBS treatment. In vitro studies revealed that pulse treatment with ALN promoted osteocalcin expression. To track the target cells of N-BPs, we applied fluorescence-labeled ALN (F-ALN) in vivo and in vitro. F-ALN was taken into bone-forming cells both in vivo and in vitro. This intracellular uptake was inhibited by endocytosis inhibitors. Furthermore, the endocytosis inhibitor dansylcadaverine (DC) suppressed ALN-stimulated osteoblastic differentiation in vitro and it suppressed the increase in alkaline phosphatase-positive bone-forming cells and subsequent bone formation in vivo. DC also blocked the inhibition of Rap1A prenylation by ALN in the osteoblastic cells. These data suggest that local application of ALN promotes bone formation by stimulating proliferation and differentiation of bone-forming cells as well as inhibiting osteoclast function. These effects may occur through endocytic incorporation of ALN and subsequent inhibition of protein prenylation.
Collapse
Affiliation(s)
- Koichiro Komatsu
- Departments of Pharmacology Orthodontics, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan Transcriptome Research Group, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-ku, Sapporo 060-8586, Japan Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Chiyoda-ku, Tokyo 113-8510, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Barthwal MK, Anzinger JJ, Xu Q, Bohnacker T, Wymann MP, Kruth HS. Fluid-phase pinocytosis of native low density lipoprotein promotes murine M-CSF differentiated macrophage foam cell formation. PLoS One 2013; 8:e58054. [PMID: 23536783 PMCID: PMC3594233 DOI: 10.1371/journal.pone.0058054] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Accepted: 01/31/2013] [Indexed: 12/21/2022] Open
Abstract
During atherosclerosis, low-density lipoprotein (LDL)-derived cholesterol accumulates in macrophages to form foam cells. Macrophage uptake of LDL promotes foam cell formation but the mechanism mediating this process is not clear. The present study investigates the mechanism of LDL uptake for macrophage colony-stimulating factor (M-CSF)-differentiated murine bone marrow-derived macrophages. LDL receptor-null (LDLR−/−) macrophages incubated with LDL showed non-saturable accumulation of cholesterol that did not down-regulate for the 24 h examined. Incubation of LDLR−/− macrophages with increasing concentrations of 125I-LDL showed non-saturable macrophage LDL uptake. A 20-fold excess of unlabeled LDL had no effect on 125I-LDL uptake by wild-type macrophages and genetic deletion of the macrophage scavenger receptors CD36 and SRA did not affect 125I-LDL uptake, showing that LDL uptake occurred by fluid-phase pinocytosis independently of receptors. Cholesterol accumulation was inhibited approximately 50% in wild-type and LDLR−/− mice treated with LY294002 or wortmannin, inhibitors of all classes of phosphoinositide 3-kinases (PI3K). Time-lapse, phase-contrast microscopy showed that macropinocytosis, an important fluid-phase uptake pathway in macrophages, was blocked almost completely by PI3K inhibition with wortmannin. Pharmacological inhibition of the class I PI3K isoforms alpha, beta, gamma or delta did not affect macrophage LDL-derived cholesterol accumulation or macropinocytosis. Furthermore, macrophages from mice expressing kinase-dead class I PI3K beta, gamma or delta isoforms showed no decrease in cholesterol accumulation or macropinocytosis when compared with wild-type macrophages. Thus, non-class I PI3K isoforms mediated macropinocytosis in these macrophages. Further characterization of the components necessary for LDL uptake, cholesterol accumulation, and macropinocytosis identified dynamin, microtubules, actin, and vacuolar type H(+)-ATPase as contributing to uptake. However, Pak1, Rac1, and Src-family kinases, which mediate fluid-phase pinocytosis in certain other cell types, were unnecessary. In conclusion, our findings provide evidence that targeting those components mediating macrophage macropinocytosis with inhibitors may be an effective strategy to limit macrophage accumulation of LDL-derived cholesterol in arteries.
Collapse
Affiliation(s)
- Manoj K. Barthwal
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joshua J. Anzinger
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Qing Xu
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas Bohnacker
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Howard S. Kruth
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
89
|
Abstract
Although effective in treating an array of neurological disorders, antipsychotics are associated with deleterious metabolic side effects. Through high-throughput screening, we previously identified phenothiazine antipsychotics as modulators of the human insulin promoter. Here, we extended our initial finding to structurally diverse typical and atypical antipsychotics. We then identified the transforming growth factor beta (TGFβ) pathway as being involved in the effect of antipsychotics on the insulin promoter, finding that antipsychotics activated SMAD3, a downstream effector of the TGFβ pathway, through a receptor distinct from the TGFβ receptor family and known neurotransmitter receptor targets of antipsychotics. Of note, antipsychotics that do not cause metabolic side effects did not activate SMAD3. In vivo relevance was demonstrated by reanalysis of gene expression data from human brains treated with antipsychotics, which showed altered expression of SMAD3 responsive genes. This work raises the possibility that antipsychotics could be designed that retain beneficial CNS activity while lacking deleterious metabolic side effects.
Collapse
Affiliation(s)
- T. Cohen
- Sanford Children’s Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA,Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA
| | | | - F. Levine
- Sanford Children’s Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA,Address correspondence to: Dr. Fred Levine, Sanford Children’s Health Research Center, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd, La Jolla, CA 92037, USA.
| |
Collapse
|
90
|
Harper CB, Popoff MR, McCluskey A, Robinson PJ, Meunier FA. Targeting membrane trafficking in infection prophylaxis: dynamin inhibitors. Trends Cell Biol 2013; 23:90-101. [DOI: 10.1016/j.tcb.2012.10.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 10/11/2012] [Accepted: 10/11/2012] [Indexed: 12/01/2022]
|
91
|
Lim J, Clements MA, Dobson J. Delivery of short interfering ribonucleic acid-complexed magnetic nanoparticles in an oscillating field occurs via caveolae-mediated endocytosis. PLoS One 2012; 7:e51350. [PMID: 23236481 PMCID: PMC3517400 DOI: 10.1371/journal.pone.0051350] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 11/01/2012] [Indexed: 11/25/2022] Open
Abstract
Gene delivery technologies to introduce foreign genes into highly differentiated mammalian cells have improved significantly over the last few decades. Relatively new techniques such as magnetic nanoparticle-based gene transfection technology are showing great promise in terms of its high transfection efficiency and wide-ranging research applications. We have developed a novel gene delivery technique, which uses magnetic nanoparticles moving under the influence of an oscillating magnetic array. Herein we successfully introduced short interfering RNA (siRNA) against green fluorescent protein (GFP) or actin into stably-transfected GFP-HeLa cells or wild-type HeLa and rat aortic smooth muscle cells, respectively. This gene silencing technique occurred in a dose- and cell density- dependent manner, as reflected using fluorescence intensity and adhesion assays. Furthermore, using endocytosis inhibitors, we established that these magnetic nanoparticle-nucleic acid complexes, moving across the cell surface under the influence of an oscillating magnet array, enters into the cells via the caveolae-mediated endocytic pathway.
Collapse
Affiliation(s)
- Jenson Lim
- nanoTherics Limited, Med IC4, Keele University Science and Business Park, Newcastle under Lyme, Staffordshire, United Kingdom
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- * E-mail:
| | - Michael A. Clements
- nanoTherics Limited, Med IC4, Keele University Science and Business Park, Newcastle under Lyme, Staffordshire, United Kingdom
| | - Jon Dobson
- J. Crayton Pruitt Family Department of Biomedical Engineering, Department of Materials Science and Engineering, and Institute for Cell Engineering and Regenerative Medicine (ICERM), University of Florida, Gainesville, Florida, United States of America
- Institute for Science and Technology in Medicine, Keele University, Stoke-On-Trent, Staffordshire, United Kingdom
| |
Collapse
|
92
|
Riva M, Källberg E, Björk P, Hancz D, Vogl T, Roth J, Ivars F, Leanderson T. Induction of nuclear factor-κB responses by the S100A9 protein is Toll-like receptor-4-dependent. Immunology 2012; 137:172-82. [PMID: 22804476 DOI: 10.1111/j.1365-2567.2012.03619.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Interactions between danger-associated molecular patterns (DAMP) and pathogen-associated molecular patterns (PAMP) and pattern recognition receptors such as Toll-like receptors (TLRs) are critical for the regulation of the inflammatory process via activation of nuclear factor-κB (NF-κB) and cytokine secretion. In this report, we investigated the capacity of lipopolysaccharide (LPS) -free S100A9 (DAMP) protein to activate human and mouse cells compared with lipoprotein-free LPS (PAMP). First, we showed that LPS and S100A9 were able to increase NF-κB activity followed by increased cytokine and nitric oxide (NO) secretion both in human THP-1 cells and in mouse bone marrow-derived dendritic cells. Surprisingly, although S100A9 triggered a weaker cytokine response than LPS, we found that S100A9 more potently induced IκBα degradation and hence NF-κB activation. Both the S100A9-induced response and the LPS-induced response were completely absent in TLR4 knockout mice, whereas it was only slightly affected in RAGE knockout mice. Also, we showed that LPS and S100A9 NF-κB induction were strongly reduced in the presence of specific inhibitors of TLR-signalling. Chloroquine reduced S100A9 but not LPS signalling, indicating that S100A9 may need to be internalized to be fully active as a TLR4 inducer. This was confirmed using A488-labelled S100A9 that was internalized in THP-1 cells, showing a raise in fluorescence after 30 min at 37°. Chloroquine treatment significantly reduced the fluorescence. In summary, our data indicate that both human and mouse S100A9 are TLR4 agonists. Importantly, S100A9 induced stronger NF-κB activation albeit weaker cytokine secretion than LPS, suggesting that S100A9 and LPS activated NF-κB in a qualitatively distinct manner.
Collapse
Affiliation(s)
- Matteo Riva
- Immunology Group, Lund University, Lund, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Differential regulation of Smad3 and of the type II transforming growth factor-β receptor in mitosis: implications for signaling. PLoS One 2012; 7:e43459. [PMID: 22927969 PMCID: PMC3425481 DOI: 10.1371/journal.pone.0043459] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 07/24/2012] [Indexed: 01/17/2023] Open
Abstract
The response to transforming growth factor-β (TGF-β) depends on cellular context. This context is changed in mitosis through selective inhibition of vesicle trafficking, reduction in cell volume and the activation of mitotic kinases. We hypothesized that these alterations in cell context may induce a differential regulation of Smads and TGF-β receptors. We tested this hypothesis in mesenchymal-like ovarian cancer cells, arrested (or not) in mitosis with 2-methoxyestradiol (2ME2). In mitosis, without TGF-β stimulation, Smad3 was phosphorylated at the C-terminus and linker regions and localized to the mitotic spindle. Phosphorylated Smad3 interacted with the negative regulators of Smad signaling, Smurf2 and Ski, and failed to induce a transcriptional response. Moreover, in cells arrested in mitosis, Smad3 levels were progressively reduced. These phosphorylations and reduction in the levels of Smad3 depended on ERK activation and Mps1 kinase activity, and were abrogated by increasing the volume of cells arrested in mitosis with hypotonic medium. Furthermore, an Mps1-dependent phosphorylation of GFP-Smad3 was also observed upon its over-expression in interphase cells, suggesting a mechanism of negative regulation which counters increases in Smad3 concentration. Arrest in mitosis also induced a block in the clathrin-mediated endocytosis of the type II TGF-β receptor (TβRII). Moreover, following the stimulation of mitotic cells with TGF-β, the proteasome-mediated attenuation of TGF-β receptor activity, the degradation and clearance of TβRII from the plasma membrane, and the clearance of the TGF-β ligand from the medium were compromised, and the C-terminus phosphorylation of Smad3 was prolonged. We propose that the reduction in Smad3 levels, its linker phosphorylation, and its association with negative regulators (observed in mitosis prior to ligand stimulation) represent a signal attenuating mechanism. This mechanism is balanced by the retention of active TGF-β receptors at the plasma membrane. Together, both mechanisms allow for a regulated cellular response to TGF-β stimuli in mitosis.
Collapse
|
94
|
Shapira KE, Gross A, Ehrlich M, Henis YI. Coated pit-mediated endocytosis of the type I transforming growth factor-β (TGF-β) receptor depends on a di-leucine family signal and is not required for signaling. J Biol Chem 2012; 287:26876-89. [PMID: 22707720 DOI: 10.1074/jbc.m112.362848] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The roles of transforming growth factor-β (TGF-β) receptor endocytosis in signaling have been investigated in numerous studies, mainly through the use of endocytosis inhibitory treatments, yielding conflicting results. Two potential sources for these discrepancies were the pleiotropic effects of a general blockade of specific internalization pathways and the scarce information on the regulation of the endocytosis of the signal-transducing type I TGF-β receptor (TβRI). Here, we employed extracellularly tagged myc-TβRI (wild type, truncation mutants, and a series of endocytosis-defective and endocytosis-enhanced mutants) to directly investigate the relationship between TβRI endocytosis and signaling. Our findings indicate that TβRI is targeted for constitutive clathrin-mediated endocytosis via a di-leucine (Leu(180)-Ile(181)) signal and an acidic cluster motif. Using Smad-dependent transcriptional activation assays and following Smad2/3 nuclear translocation in response to TGF-β stimulation, we show that TβRI endocytosis is dispensable for TGF-β signaling and may play a role in signal termination. Alanine replacement of Leu(180)-Ile(181) led to partial constitutive activation of TβRI, resulting in part from its retention at the plasma membrane and in part from potential alterations of TβRI regulatory interactions in the vicinity of the mutated residues.
Collapse
Affiliation(s)
- Keren E Shapira
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | |
Collapse
|
95
|
Targeted disruption of core 1 β1,3-galactosyltransferase (C1galt1) induces apical endocytic trafficking in human corneal keratinocytes. PLoS One 2012; 7:e36628. [PMID: 22574202 PMCID: PMC3344913 DOI: 10.1371/journal.pone.0036628] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 04/10/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Exposed mucosal surfaces limit constitutive endocytosis under physiological conditions to prevent uptake of macromolecules and pathogens and, therefore, cellular damage. It is now accepted that cell surface mucins, a group of high molecular weight glycoproteins on the epithelial glycocalyx, defined by their extensive O-glycosylation, play a major role in maintaining barrier function in these surfaces, but the precise mechanisms are unclear. METHODOLOGY/PRINCIPAL FINDINGS In this work, we utilized a stable tetracycline-inducible RNA interfering system targeting the core 1 ß1,3-galactosyltransferase (C1galt1 or T-synthase), a critical galactosyltransferase required for the synthesis of core 1 O-glycans, to explore the role of mucin-type carbohydrates in apical endocytic trafficking in human corneal keratinocytes. Using cell surface biotinylation and subcellular fractionation, we found increased accumulation of plasma membrane protein in endosomes after C1galt1 depletion. Confocal laser scanning microscopy and fluorometry revealed increased translocation of negatively charged fluorescent nanospheres after C1galt1 knockdown sustained by an active transport process and largely independent of apical intercellular junctions. Internalization of nanospheres could be blocked by dynasore, nocodazole, chlorpromazine, and hyperosmotic sucrose, suggesting a mechanism for clathrin-coated pit budding and vesicular trafficking. This possibility was supported by experiments showing nanosphere colocalization with clathrin heavy chain in the cytoplasm. CONCLUSIONS/SIGNIFICANCE Together, the data suggest that core 1 O-glycans contribute to maintenance of apical barrier function on exposed mucosal surfaces by preventing clathrin-mediated endocytosis.
Collapse
|
96
|
Ma CIJ, Martin C, Ma Z, Hafiane A, Dai M, Lebrun JJ, Kiss RS. Engulfment protein GULP is regulator of transforming growth factor-β response in ovarian cells. J Biol Chem 2012; 287:20636-51. [PMID: 22451657 DOI: 10.1074/jbc.m111.314997] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor β (TGF-β) is a key regulatory molecule with pleiotropic effects on cell growth, migration, and invasion. As a result, impairment of proper TGF-β signaling is central to tumorigenesis and metastasis. The TGF-β receptor V (TGFBRV or LRP1) has been shown to be responsible for TGF-β-mediated cell growth inhibition in Chinese hamster ovary (CHO) cells. The LRP1 adapter protein GULP mediates internalization of the various LRP1-specific ligands, and we hypothesize that GULP acts as a novel regulator of TGF-β signaling in ovarian cells. CHO cells that overexpress exogenous GULP (FL) demonstrate enhancement in growth inhibition, migration, and invasion from TGF-β treatment, whereas cells that lack GULP (AS) show impairment of growth inhibition and decreased migration and invasion. The enhanced TGF-β response in FL cells was confirmed by a prolonged TGF-β-induced SMAD3 phosphorylation, whereas a shortening of the phosphorylation event is observed in AS cells. Mechanistically, the presence of GULP retains the TGF-β in a signaling-competent early endosome for enhanced signaling. To address this mechanism in a physiological setting, TGF-β insensitive ovarian adenocarcinoma cells (HEY) have a very low GULP expression level, similar to the observation made in a wide selection of human ovarian adenocarcinomas. Transfection of GULP into the HEY cells restored the TGF-β responsiveness, as measured by SMAD3 phosphorylation and impairment of cell growth. Because GULP expression positively regulates TGF-β signaling leading to growth inhibition, this may represent an attractive target to achieve TGF-β responsiveness in ovarian cells.
Collapse
Affiliation(s)
- Cheng-I J Ma
- Department of Medicine, McGill University, Montreal, Quebec H3A 1A1, Canada
| | | | | | | | | | | | | |
Collapse
|
97
|
Morcavallo A, Genua M, Palummo A, Kletvikova E, Jiracek J, Brzozowski AM, Iozzo RV, Belfiore A, Morrione A. Insulin and insulin-like growth factor II differentially regulate endocytic sorting and stability of insulin receptor isoform A. J Biol Chem 2012; 287:11422-36. [PMID: 22318726 DOI: 10.1074/jbc.m111.252478] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The insulin receptor isoform A (IR-A) binds both insulin and insulin-like growth factor (IGF)-II, although the affinity for IGF-II is 3-10-fold lower than insulin depending on a cell and tissue context. Notably, in mouse embryonic fibroblasts lacking the IGF-IR and expressing solely the IR-A (R-/IR-A), IGF-II is a more potent mitogen than insulin. As receptor endocytosis and degradation provide spatial and temporal regulation of signaling events, we hypothesized that insulin and IGF-II could affect IR-A biological responses by differentially regulating IR-A trafficking. Using R-/IR-A cells, we discovered that insulin evoked significant IR-A internalization, a process modestly affected by IGF-II. However, the differential internalization was not due to IR-A ubiquitination. Notably, prolonged stimulation of R-/IR-A cells with insulin, but not with IGF-II, targeted the receptor to a degradative pathway. Similarly, the docking protein insulin receptor substrate 1 (IRS-1) was down-regulated after prolonged insulin but not IGF-II exposure. Similar results were also obtained in experiments using [NMeTyr(B26)]-insulin, an insulin analog with IR-A binding affinity similar to IGF-II. Finally, we discovered that IR-A was internalized through clathrin-dependent and -independent pathways, which differentially regulated the activation of downstream effectors. Collectively, our results suggest that a lower affinity of IGF-II for the IR-A promotes lower IR-A phosphorylation and activation of early downstream effectors vis à vis insulin but may protect IR-A and IRS-1 from down-regulation thereby evoking sustained and robust mitogenic stimuli.
Collapse
Affiliation(s)
- Alaide Morcavallo
- Department of Urology and Endocrine Mechanisms and Hormone Action Program, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Surface immobilization of bone morphogenetic protein 2 via a self-assembled monolayer formation induces cell differentiation. Acta Biomater 2012; 8:772-80. [PMID: 22040684 DOI: 10.1016/j.actbio.2011.10.019] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 10/12/2011] [Accepted: 10/13/2011] [Indexed: 11/23/2022]
Abstract
Bone extracellular matrix consists of a network of proteins in which growth factors, like bone morphogenetic protein 2 (BMP-2), are embedded and released upon matrix turnover and degradation. Recombinant human (rh)BMP-2 shows promise in enhancing bone fracture repair, although issues regarding finding a suitable delivery system still limit its extensive clinical use. The aim of this study is to determine which cell activities are triggered by the presentation of immobilized rhBMP-2. For this purpose gold surfaces were first decorated with a self-assembled monolayer consisting of a hetero-bifunctional linker. rhBMP-2 was covalently bound to the surfaces via this linker and used to investigate the cellular responses of C2C12 myoblasts. We show that covalently immobilized rhBMP-2 (iBMP-2) initiates short-term signaling events. Using a BMP-responsive reporter gene assay and western blotting to monitor phosphorylation of Smad1/5/8 we prove that iBMP-2 activates BMP-dependent signal transduction. Furthermore, we demonstrate that iBMP-2 suppresses myotube formation and promotes the osteoblast phenotype in C2C12 cells. The bioactivity of surface-bound rhBMP-2 presented in this study is not due to its release into the medium. As such, our simple approach paves the way for the controlled local presentation of immobilized growth factors, limiting degradation while still maintaining biological activity.
Collapse
|
99
|
Meyer zu Hörste M, Ströher E, Berchner-Pfannschmidt U, Schmitz-Spanke S, Pink M, Göthert JR, Fischer JW, Gulbins E, Eckstein AK. A novel mechanism involved in the pathogenesis of Graves ophthalmopathy (GO): clathrin is a possible targeting molecule for inhibiting local immune response in the orbit. J Clin Endocrinol Metab 2011; 96:E1727-36. [PMID: 21917865 DOI: 10.1210/jc.2011-1156] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Excessive orbital fibroblast (OF) proliferation and extracellular matrix production, as well as inflammation resulting in the expansion and remodeling of orbital tissue, are characteristic of Graves ophthalmopathy (GO). Our aim was to analyze and inhibit signaling pathways in resident OF that are involved in GO. METHODS/MAIN OUTCOME MEASURES: Primary human OF were obtained from 12 patients with active, severe GO and from 12 healthy control subjects. The cells were characterized by immunofluorescence assay and flow cytometry. Tyrosine phosphorylation of cellular proteins was determined by Western blot techniques, immunoprecipitation, and protein identity with mass spectrometry. Cell proliferation was determined by 5-bromo-2-deoxyuridine incorporation, hyaluronan (HA) production was assessed by a HA-binding protein based assay, and intracellular reactive oxygen species (ROS) were determined by the dichlorofluorescein assay. Clathrin heavy-chain (CHC) expression was inhibited with small interfering RNA technology. RESULTS Tyrosine phosphorylation of CHC is constitutively increased in vitro in GO-derived OF, independent of serum or other stimulating factors. The proliferative and biosynthetic capabilities (production of HA, ROS) of GO-derived OF are significantly higher than those of OF from healthy control subjects. Down-regulation of CHC expression leads to a normalization of pathologically increased proliferation and production of HA and ROS in GO-derived OFs in vitro. CONCLUSIONS Our findings strongly suggest that clathrin and clathrin-mediated signaling pathways are involved in the inflammatory signal transduction of OF in GO. With the identification of clathrin, we report a new potential targeting molecule for specific pharmacological inhibition of the local inflammatory response characteristic of GO.
Collapse
Affiliation(s)
- Melissa Meyer zu Hörste
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Effects of transport inhibitors on the cellular uptake of carboxylated polystyrene nanoparticles in different cell lines. PLoS One 2011; 6:e24438. [PMID: 21949717 PMCID: PMC3176276 DOI: 10.1371/journal.pone.0024438] [Citation(s) in RCA: 325] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 08/10/2011] [Indexed: 12/31/2022] Open
Abstract
Nanotechnology is expected to play a vital role in the rapidly developing field of nanomedicine, creating innovative solutions and therapies for currently untreatable diseases, and providing new tools for various biomedical applications, such as drug delivery and gene therapy. In order to optimize the efficacy of nanoparticle (NP) delivery to cells, it is necessary to understand the mechanisms by which NPs are internalized by cells, as this will likely determine their ultimate sub-cellular fate and localisation. Here we have used pharmacological inhibitors of some of the major endocytic pathways to investigate nanoparticle uptake mechanisms in a range of representative human cell lines, including HeLa (cervical cancer), A549 (lung carcinoma) and 1321N1 (brain astrocytoma). Chlorpromazine and genistein were used to inhibit clathrin and caveolin mediated endocytosis, respectively. Cytochalasin A and nocodazole were used to inhibit, respectively, the polymerisation of actin and microtubule cytoskeleton. Uptake experiments were performed systematically across the different cell lines, using carboxylated polystyrene NPs of 40 nm and 200 nm diameters, as model NPs of sizes comparable to typical endocytic cargoes. The results clearly indicated that, in all cases and cell types, NPs entered cells via active energy dependent processes. NP uptake in HeLa and 1321N1 cells was strongly affected by actin depolymerisation, while A549 cells showed a stronger inhibition of NP uptake (in comparison to the other cell types) after microtubule disruption and treatment with genistein. A strong reduction of NP uptake was observed after chlorpromazine treatment only in the case of 1321N1 cells. These outcomes suggested that the same NP might exploit different uptake mechanisms to enter different cell types.
Collapse
|