1
|
Wang Y, Ni M, Huang M, Xing L, Liu X, Jia F, Huang Y. Regulation of nanoparticle exocytosis direction via receptors transfer: A novel strategy to enhance therapeutic efficacy of semaglutide. Int J Pharm 2025; 674:125439. [PMID: 40064382 DOI: 10.1016/j.ijpharm.2025.125439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
Coumaric acid (CA) is a typical nutrient required in relatively high quantities by the body. It has been proved CA could specifically bind to monocarboxylate Transporter-1 (MCT-1) receptors, a transporter protein expressed on the surface of intestinal epithelial cells, to facilitate its cellular uptake. Although our preliminary research demonstrated semaglutide (SEM) loaded CA modified nanoparticles (SEM@CNP) could improve the absorption of SEM to some extent, the oral bioavailability still remained suboptimal owing to the lysosomal degradation. To address this issue, INF-7 (peptide chain GLFEAIEGFIENGWEGMIDGWYG) and chloroquine (CQ), two lysosomal escape agents (LEAs) with different mechanisms of action, were incorporated with SEM@CNP for oral delivery (SEM@CNP + INF-7, SEM@CNP + CQ). In type II diabetes mice models, SEM@CNP + CQ effectively inhibited postprandial glucose rise with a relative pharmacological bioavailability of 20.63 ± 2.99 %, 1.73 times higher than SEM@CNP (11.90 ± 4.56 %). Mechanistic studies revealed that: 1) after adding LEAs, the exocytosis preference of nanoparticles was altered, tending towards basolateral exocytosis apparently. Regulated exocytosis directionality was linked to the spatial redistribution of MCT-1 receptors. 2) among the two LEAs, CQ demonstrated superior efficacy compared to INF-7. This superiority was attributed to the earlier onset of action and more pronounced degree of membrane disruption induced by CQ. This research provided new insights for the design of oral delivery systems for peptidic drugs.
Collapse
Affiliation(s)
- Yating Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Mingjie Ni
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Minyi Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Liyun Xing
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xi Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Fuya Jia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Ji X, Wang X, Dong Q, Li W, Zhou N, Yue X, Zhao D, Yang X. CDCP1 knockdown suppresses PDGFRβ/AKT pathway-mediated vascular smooth muscle cell proliferation by inhibiting PDGFRβ endocytosis. PeerJ 2025; 13:e19114. [PMID: 40256729 PMCID: PMC12007496 DOI: 10.7717/peerj.19114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 02/13/2025] [Indexed: 04/22/2025] Open
Abstract
CUB domain-containing protein 1 (CDCP1) is a type of cell surface glycoprotein that has been identified as being capable of regulating cell anchorage, migration, proliferation, and differentiation. However, the contributions of CDCP1 in intimal hyperplasia, specifically regarding the proliferation and migration of vascular smooth muscle cells (VSMC), are unclear. In this study, we analyzed CDCP1 expression on intimal hyperplasia through a focal carotid stenosis model in vivo. In vitro, we cultured mouse VSMCs and stimulated them with 20 ng/mL platelet-derived growth factor BB (PDGF-BB) for 24 h. Western blot analysis was performed to detect the expression of CDCP1 in the cells. Next, we knocked down the expression of CDCP1 in VSMCs and assessed its effects on cell proliferation and migration using CCK8 assays, EDU+ assay, and wound healing experiments. We then performed RNA-Seq analysis on the CDCP1-knockdown VSMCs. Based on the sequencing results, we utilized western blotting to investigate the impact of CDCP1 knockdown on the AKT signaling pathway. Additionally, we validated the interactions between Platelet-derived growth factor receptor (PDGFR)β with NEDD4, clathrin, and Rab5 using immunofluorescence and co-immunoprecipitation assays. The results discovered that CDCP1 levels were activated in the intimal hyperplasia tissues in vivo. CDCP1 knockdown significantly attenuated mouse VSMC proliferation and migration induced by PDGF-BB in vitro. Based on the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of the differentially expressed proteins obtained from RNA-sequencing, we found that the knockdown of CDCP1 is related to the "PI3K-AKT signaling pathway", "ubiquitin-mediated proteolysis", and "endocytosis" pathways. The subsequent experiments demonstrated that CDCP1 knockdown inhibited AKT signaling pathway. CDCP1 knockdown promoted the binding of PDGFRβ and NEDD4, and PDGFRβ ubiquitin. Moreover, CDCP1 knockdown attenuated the binding of PDGFRβ to clathrin and Rab5. These data reveal that the absence of CDCP1 may enhance the binding of PDGFR to NEDD4 and promote the ubiquitination of PDGFR, thereby regulating the AKT signaling pathway and intimal hyperplasia.
Collapse
MESH Headings
- Animals
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Cell Proliferation/genetics
- Mice
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- Endocytosis/genetics
- Myocytes, Smooth Muscle/metabolism
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Movement
- Gene Knockdown Techniques
- Becaplermin/pharmacology
- Male
- Cells, Cultured
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Xin Ji
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Collaborative Innovation Center for Eco-Environment, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, China
- Department of Clinical Laboratory, Southern University of Science and Technology Hospital, Shenzhen, Guangdong Province, China
| | - Xin Wang
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Qianqian Dong
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Wanqiu Li
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Ning Zhou
- Department of Clinical Laboratory, Southern University of Science and Technology Hospital, Shenzhen, Guangdong Province, China
| | - Xiaole Yue
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Dandan Zhao
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xiaolong Yang
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Collaborative Innovation Center for Eco-Environment, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
3
|
Tang M, Yarragudi SB, Pan P, Yang K, Kanamala M, Wu Z. Effect of size and pH-sensitivity of liposomes on cellular uptake pathways and pharmacokinetics of encapsulated gemcitabine. J Liposome Res 2025; 35:44-54. [PMID: 39126197 DOI: 10.1080/08982104.2024.2389969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
To enhance cytoplasmic delivery efficiency, pH-sensitive liposomes (PSL) have been proposed as a novel strategy. To facilitate clinical translation, this study aims to understand the impact of both size and pH-sensitivity on cellular uptake pathways, intracellular trafficking and pharmacokinetics of liposomes. The large liposomes (130-160 nm) were prepared using thin-film hydration method, while small liposomes (∼60 nm) were fabricated using microfluidics, for both PSL and non-pH-sensitive liposomes (NPSL). Cellular uptake pathways and intracellular trafficking was investigated through confocal imaging with aid of various endocytosis inhibitors. Intracellular gemcitabine delivery by various liposomal formulations was quantified using HPLC, and the cytotoxicity was assessed via cell viability assays. Pharmacokinetics of gemcitabine loaded in various liposomes was evaluated in rats following intravenous administration. Larger liposomes had a higher loading capacity for hydrophilic gemcitabine (7% vs 4%). Small PSL exhibited superior cellular uptake compared to large PSL or NPSLs. Moreover, the alkalization of endosomes significantly attenuated the cellular uptake of PSL. Large liposomes (PSL and NPSL) predominantly entered cells via clathrin-dependent pathway, whereas small liposomes partially utilized caveolae-dependent pathway. However, the long circulation of the liposomes, as measured by the encapsulated gemcitabine, was compromised by both pH-sensitivity and size reduction (9.5 h vs 5.3 h). Despite this drawback, our results indicate that small PSL holds promise as vectors for the next generation of liposomal nanomedicine, owing to their superior cytoplasmic delivery efficiency.
Collapse
Affiliation(s)
- Mingtan Tang
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Sasi Bhushan Yarragudi
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Patrick Pan
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kaiyun Yang
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Manju Kanamala
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Zimei Wu
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
4
|
Tai SB, Huang CY, Chung CL, Sung PJ, Wen ZH, Chen CL. Prodigiosin Inhibits Transforming Growth Factor β Signaling by Interfering Receptor Recycling and Subcellular Translocation in Epithelial Cells. Mol Pharmacol 2024; 105:286-300. [PMID: 38278554 DOI: 10.1124/molpharm.123.000776] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 01/28/2024] Open
Abstract
Prodigiosin (PG) is a naturally occurring polypyrrole red pigment produced by numerous microorganisms including some Serratia and Streptomyces strains. PG has exhibited promising anticancer activity; however, the molecular mechanisms of action of PG on malignant cells remain ambiguous. Transforming growth factor-β (TGF-β) is a multifunctional cytokine that governs a wide array of cellular processes in development and tissue homeostasis. Malfunctions of TGF-β signaling are associated with numerous human cancers. Emerging evidence underscores the significance of internalized TGF-β receptors and their intracellular trafficking in initiating signaling cascades. In this study, we identified PG as a potent inhibitor of the TGF-β pathway. PG blocked TGF-β signaling by targeting multiple sites of this pathway, including facilitating the sequestering of TGF-β receptors in the cytoplasm by impeding the recycling of type II TGF-β receptors to the cell surface. Additionally, PG prompts a reduction in the abundance of receptors on the cell surface through the disruption of the receptor glycosylation. In human Caucasian lung carcinoma cells and human hepatocellular cancer cell line cells, nanomolar concentrations of PG substantially diminish TGF-β-triggered phosphorylation of Smad2 protein. This attenuation is further reflected in the suppression of downstream target gene expression, including those encoding fibronectin, plasminogen activator inhibitor-1, and N-cadherin. SIGNIFICANCE STATEMENT: Prodigiosin (PG) emerges from this study as a potent TGF-β pathway inhibitor, disrupting receptor trafficking and glycosylation and reducing TGF-β signaling and downstream gene expression. These findings not only shed light on PG's potential therapeutic role but also present a captivating avenue towards future anti-TGF-β strategies.
Collapse
Affiliation(s)
- Shun-Ban Tai
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Yin Huang
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Ling Chung
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ping-Jyun Sung
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zhi-Hong Wen
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Lin Chen
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Abboud D, Abboud C, Inoue A, Twizere JC, Hanson J. Basal interaction of the orphan receptor GPR101 with arrestins leads to constitutive internalization. Biochem Pharmacol 2024; 220:116013. [PMID: 38151077 DOI: 10.1016/j.bcp.2023.116013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 12/29/2023]
Abstract
GPR101 is an orphan G protein-coupled receptor that promotes growth hormone secretion in the pituitary. The microduplication of the GPR101 gene has been linked with the X-linked acrogigantism, or X-LAG, syndrome. This disease is characterized by excessive growth hormone secretion and abnormal rapid growth beginning early in life. Mechanistically, GPR101 induces growth hormone secretion through constitutive activation of multiple heterotrimeric G proteins. However, the full scope of GPR101 signaling remains largely elusive. Herein, we investigated the association of GPR101 to multiple transducers and uncovered an important basal interaction with Arrestin 2 (β-arrestin 1) and Arrestin 3 (β-arrestin 2). By using a GPR101 mutant lacking the C-terminus and cell lines with an Arrestin 2/3 null background, we show that the arrestin association leads to constitutive clathrin- and dynamin-mediated GPR101 internalization. To further highlight GPR101 intracellular fate, we assessed the colocalization of GPR101 with Rab protein markers. Internalized GPR101 was mainly colocalized with the early endosome markers, Rab5 and EEA-1, and to a lesser degree with the late endosome marker Rab7. However, GPR101 was not colocalized with the recycling endosome marker Rab11. These findings show that the basal arrestin recruitment by GPR101 C-terminal tail drives the receptor constitutive clathrin-mediated internalization. Intracellularly, GPR101 concentrates in the endosomal compartment and is degraded through the lysosomal pathway. In conclusion, we uncovered a constitutive intracellular trafficking of GPR101 that potentially represents an important layer of regulation of its signaling and function.
Collapse
Affiliation(s)
- Dayana Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | - Clauda Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Jean-Claude Twizere
- Laboratory of Viral Interactomes, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | - Julien Hanson
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium; Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), University of Liege, Liege, Belgium.
| |
Collapse
|
6
|
Chaudhary R, Goodman LS, Wang S, Asimakopoulos A, Weiskirchen R, Dooley S, Ehrlich M, Henis YI. Cholesterol modulates type I/II TGF-β receptor complexes and alters the balance between Smad and Akt signaling in hepatocytes. Commun Biol 2024; 7:8. [PMID: 38168942 PMCID: PMC10761706 DOI: 10.1038/s42003-023-05654-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Cholesterol mediates membrane compartmentalization, affecting signaling via differential distribution of receptors and signaling mediators. While excessive cholesterol and aberrant transforming growth factor-β (TGF-β) signaling characterize multiple liver diseases, their linkage to canonical vs. non-canonical TGF-β signaling remained unclear. Here, we subjected murine hepatocytes to cholesterol depletion (CD) or enrichment (CE), followed by biophysical studies on TGF-β receptor heterocomplex formation, and output to Smad2/3 vs. Akt pathways. Prior to ligand addition, raft-dependent preformed heteromeric receptor complexes were observed. Smad2/3 phosphorylation persisted following CD or CE. CD enhanced phospho-Akt (pAkt) formation by TGF-β or epidermal growth factor (EGF) at 5 min, while reducing it at later time points. Conversely, pAkt formation by TGF-β or EGF was inhibited by CE, suggesting a direct effect on the Akt pathway. The modulation of the balance between TGF-β signaling to Smad2/3 vs. pAkt (by TGF-β or EGF) has potential implications for hepatic diseases and malignancies.
Collapse
Affiliation(s)
- Roohi Chaudhary
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Laureen S Goodman
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Sai Wang
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, D-68167, Mannheim, Germany
| | - Anastasia Asimakopoulos
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, D-52074, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, D-52074, Aachen, Germany
| | - Steven Dooley
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, D-68167, Mannheim, Germany
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
7
|
Shrivastava P, Mahale A, Prakash Kulkarni O, Kashaw SK, Vyas SP. Targeted intracellular delivery of antitubercular bioactive(s) to Mtb infected macrophages via transferrin functionalized nanoliposomes. Int J Pharm 2023:123189. [PMID: 37391107 DOI: 10.1016/j.ijpharm.2023.123189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
The packaging of antimicrobials/chemotherapeutics into nanoliposomes can enhance their activity while minimizing toxicity. However, their use is still limited owing to inefficient/inadequate loading strategies. Several bioactive(s) which are non ionizable, and poorly aqueous soluble cannot be easily encapsulated into aqueous core of liposomes by using conventional means. Such bioactive(s) however could be encapsulated in the liposomes by forming their water soluble molecular inclusion complex with cyclodextrins. In this study, we developed Rifampicin (RIF) - 2-hydroxylpropyl-β-cyclodextrin (HP-β-CD) molecular inclusion complex. The HP-β-CD-RIF complex interaction was assessed by using computational analysis (molecular modeling). The HP-β-CD-RIF complex and Isoniazid were co-loaded in the small unilamellar vesicles (SUVs). Further, the developed system was functionalized with transferrin, a targeting moiety. Transferrin functionalized SUVs (Tf-SUVs) could preferentially deliver their payload intracellularly in the endosomal compartment of macrophages. In in vitro study on infected Raw 264.7 macrophage cells revealed that the encapsulated bioactive(s) could eradicate the pathogen more efficiently than free bioactive(s). In vivo studies further revealed that the Tf-SUVs could accumulate and maintain intracellular bioactive(s) concentrations in macrophages. The study suggests Tf-SUVs as a promising module for targeted delivery of a drug combination with improved/optimal therapeutic index and effective clinical outcomes.
Collapse
Affiliation(s)
- Priya Shrivastava
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, (M.P.), India, 470003
| | - Ashutosh Mahale
- Department of Pharmacy (Pharmacology division), Birla Institute of Technology and Science Pilani, Hyderabad Campus, Telangana, 500078, India
| | - Onkar Prakash Kulkarni
- Department of Pharmacy (Pharmacology division), Birla Institute of Technology and Science Pilani, Hyderabad Campus, Telangana, 500078, India
| | - Sushil K Kashaw
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, (M.P.), India, 470003
| | - Suresh P Vyas
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, (M.P.), India, 470003.
| |
Collapse
|
8
|
Banushi B, Joseph SR, Lum B, Lee JJ, Simpson F. Endocytosis in cancer and cancer therapy. Nat Rev Cancer 2023:10.1038/s41568-023-00574-6. [PMID: 37217781 DOI: 10.1038/s41568-023-00574-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/24/2023]
Abstract
Endocytosis is a complex process whereby cell surface proteins, lipids and fluid from the extracellular environment are packaged, sorted and internalized into cells. Endocytosis is also a mechanism of drug internalization into cells. There are multiple routes of endocytosis that determine the fate of molecules, from degradation in the lysosomes to recycling back to the plasma membrane. The overall rates of endocytosis and temporal regulation of molecules transiting through endocytic pathways are also intricately linked with signalling outcomes. This process relies on an array of factors, such as intrinsic amino acid motifs and post-translational modifications. Endocytosis is frequently disrupted in cancer. These disruptions lead to inappropriate retention of receptor tyrosine kinases on the tumour cell membrane, changes in the recycling of oncogenic molecules, defective signalling feedback loops and loss of cell polarity. In the past decade, endocytosis has emerged as a pivotal regulator of nutrient scavenging, response to and regulation of immune surveillance and tumour immune evasion, tumour metastasis and therapeutic drug delivery. This Review summarizes and integrates these advances into the understanding of endocytosis in cancer. The potential to regulate these pathways in the clinic to improve cancer therapy is also discussed.
Collapse
Affiliation(s)
- Blerida Banushi
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Shannon R Joseph
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Benedict Lum
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Jason J Lee
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| |
Collapse
|
9
|
Members of Venezuelan Equine Encephalitis complex entry into host cells by clathrin-mediated endocytosis in a pH-dependent manner. Sci Rep 2022; 12:14556. [PMID: 36008558 PMCID: PMC9411563 DOI: 10.1038/s41598-022-18846-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/22/2022] [Indexed: 01/20/2023] Open
Abstract
Pixuna virus (PIXV) and Río Negro virus (RNV) are mosquito-borne alphaviruses belonging to the Venezuelan Equine Encephalitis (VEE) complex, which includes pathogenic epizootic and enzootic subtypes responsible for life-threatening diseases in equines. Considering that the first steps in viral infection are crucial for the efficient production of new progeny, the aim of this study was to elucidate the early events of the replication cycle of these two viruses. To this end, we used chemical inhibitors and the expression of dominant-negative constructs to study the dependence of clathrin and endosomal pH on PIXV and RNV internalization mechanisms. We demonstrated that both viruses are internalized primarily via clathrin-mediated endocytosis, where the low pH in endosomes is crucial for viral replication. Contributing knowledge regarding the entry route of VEE complex members is important to understand the pathogenesis of these viruses and also to develop new antiviral strategies.
Collapse
|
10
|
Abstract
Anti-Müllerian Hormone (AMH) is a secreted glycoprotein hormone with critical roles in reproductive development and regulation. Its chemical and mechanistic similarities to members of the Transforming Growth Factor β (TGF-β) family have led to its placement within this signaling family. As a member of the TGF-β family, AMH exists as a noncovalent complex of a large N-terminal prodomain and smaller C-terminal mature signaling domain. To produce a signal, the mature domain will bind to the extracellular domains of two type I and two type II receptors which results in an intracellular SMAD signal. Interestingly, as will be discussed in this review, AMH possesses several unique characteristics which set it apart from other ligands within the TGF-β family. In particular, AMH has a dedicated type II receptor, Anti-Müllerian Hormone Receptor Type II (AMHR2), making this interaction intriguing mechanistically as well as therapeutically. Further, the prodomain of AMH has remained largely uncharacterized, despite being the largest prodomain within the family. Recent advancements in the field have provided valuable insight into the molecular mechanisms of AMH signaling, however there are still many areas of AMH signaling not understood. Herein, we will discuss what is known about the biochemistry of AMH and AMHR2, focusing on recent advances in understanding the unique characteristics of AMH signaling and the molecular mechanisms of receptor engagement.
Collapse
Affiliation(s)
- James A. Howard
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Kaitlin N. Hart
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Thomas B. Thompson
- Department of Molecular Genetics, Biochemistry, & Microbiology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
11
|
Abstract
Current therapies for pulmonary fibrosis (PF) focus on slowing disease progression and reducing functional decline in patients by dampening the activation of fibroblasts and other implicated cells. There is a need for strategies that target the essential cells and signaling pathways involved in disease pathogenesis. Monocyte-derived macrophages (Mo-Macs) are known to express profibrotic genes and are involved in the pathogenesis of PF. Our results show that engineered mannosylated albumin nanoparticles specifically targeted disease-inducing Mo-Macs, and further, that nanoparticles efficiently delivered small-interfering RNA against profibrotic cytokine tumor growth factor β1 to prevent bleomycin-induced lung fibrosis. The pathogenesis of lung fibrosis involves hyperactivation of innate and adaptive immune pathways that release inflammatory cytokines and growth factors such as tumor growth factor (TGF)β1 and induce aberrant extracellular matrix protein production. During the genesis of pulmonary fibrosis, resident alveolar macrophages are replaced by a population of newly arrived monocyte-derived interstitial macrophages that subsequently transition into alveolar macrophages (Mo-AMs). These transitioning cells initiate fibrosis by releasing profibrotic cytokines and remodeling the matrix. Here, we describe a strategy for leveraging the up-regulation of the mannose receptor CD206 in interstitial macrophages and Mo-AM to treat lung fibrosis. We engineered mannosylated albumin nanoparticles, which were found to be internalized by fibrogenic CD206+ monocyte derived macrophages (Mo-Macs). Mannosylated albumin nanoparticles incorporating TGFβ1 small-interfering RNA (siRNA) targeted the profibrotic subpopulation of CD206+ macrophages and prevented lung fibrosis. The findings point to the potential utility of mannosylated albumin nanoparticles in delivering TGFβ-siRNA into CD206+ profibrotic macrophages as an antilung fibrosis strategy.
Collapse
|
12
|
Bugatti A, Filippini F, Bardelli M, Zani A, Chiodelli P, Messali S, Caruso A, Caccuri F. SARS-CoV-2 Infects Human ACE2-Negative Endothelial Cells through an αvβ3 Integrin-Mediated Endocytosis Even in the Presence of Vaccine-Elicited Neutralizing Antibodies. Viruses 2022; 14:v14040705. [PMID: 35458435 PMCID: PMC9032829 DOI: 10.3390/v14040705] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 02/06/2023] Open
Abstract
Integrins represent a gateway of entry for many viruses and the Arg-Gly-Asp (RGD) motif is the smallest sequence necessary for proteins to bind integrins. All Severe Acute Respiratory Syndrome Virus type 2 (SARS-CoV-2) lineages own an RGD motif (aa 403–405) in their receptor binding domain (RBD). We recently showed that SARS-CoV-2 gains access into primary human lung microvascular endothelial cells (HL-mECs) lacking Angiotensin-converting enzyme 2 (ACE2) expression through this conserved RGD motif. Following its entry, SARS-CoV-2 remodels cell phenotype and promotes angiogenesis in the absence of productive viral replication. Here, we highlight the αvβ3 integrin as the main molecule responsible for SARS-CoV-2 infection of HL-mECs via a clathrin-dependent endocytosis. Indeed, pretreatment of virus with αvβ3 integrin or pretreatment of cells with a monoclonal antibody against αvβ3 integrin was found to inhibit SARS-CoV-2 entry into HL-mECs. Surprisingly, the anti-Spike antibodies evoked by vaccination were neither able to impair Spike/integrin interaction nor to prevent SARS-CoV-2 entry into HL-mECs. Our data highlight the RGD motif in the Spike protein as a functional constraint aimed to maintain the interaction of the viral envelope with integrins. At the same time, our evidences call for the need of intervention strategies aimed to neutralize the SARS-CoV-2 integrin-mediated infection of ACE2-negative cells in the vaccine era.
Collapse
Affiliation(s)
- Antonella Bugatti
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.B.); (F.F.); (M.B.); (A.Z.); (S.M.); (A.C.)
| | - Federica Filippini
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.B.); (F.F.); (M.B.); (A.Z.); (S.M.); (A.C.)
| | - Marta Bardelli
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.B.); (F.F.); (M.B.); (A.Z.); (S.M.); (A.C.)
| | - Alberto Zani
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.B.); (F.F.); (M.B.); (A.Z.); (S.M.); (A.C.)
| | - Paola Chiodelli
- Section of General Pathology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Serena Messali
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.B.); (F.F.); (M.B.); (A.Z.); (S.M.); (A.C.)
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.B.); (F.F.); (M.B.); (A.Z.); (S.M.); (A.C.)
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.B.); (F.F.); (M.B.); (A.Z.); (S.M.); (A.C.)
- Correspondence:
| |
Collapse
|
13
|
Durán-Lobato M, Álvarez-Fuentes J, Fernández-Arévalo M, Martín-Banderas L. Receptor-targeted nanoparticles modulate cannabinoid anticancer activity through delayed cell internalization. Sci Rep 2022; 12:1297. [PMID: 35079042 PMCID: PMC8789857 DOI: 10.1038/s41598-022-05301-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 12/24/2021] [Indexed: 12/15/2022] Open
Abstract
Δ9-tetrahydrocannabinol (Δ9-THC) is known for its antitumor activity and palliative effects. However, its unfavorable physicochemical and biopharmaceutical properties, including low bioavailability, psychotropic side effects and resistance mechanisms associated to dosing make mandatory the development of successful drug delivery systems. In this work, transferring (Tf) surface-modified Δ9-THC-loaded poly(lactide-co-glycolic) nanoparticles (Tf-THC-PLGA NPs) were proposed and evaluated as novel THC-based anticancer therapy. Furthermore, in order to assess the interaction of both the nanocarrier and the loaded drug with cancer cells, a double-fluorescent strategy was applied, including the chemical conjugation of a dye to the nanoparticle polymer along with the encapsulation of either a lipophilic or a hydrophilic dye. Tf-THC PLGA NPs exerted a cell viability decreased down to 17% vs. 88% of plain nanoparticles, while their internalization was significantly slower than plain nanoparticles. Uptake studies in the presence of inhibitors indicated that the nanoparticles were internalized through cholesterol-associated and clathrin-mediated mechanisms. Overall, Tf-modification of PLGA NPs showed to be a highly promising approach for Δ9-THC-based antitumor therapies, potentially maximizing the amount of drug released in a sustained manner at the surface of cells bearing cannabinoid receptors.
Collapse
Affiliation(s)
- Matilde Durán-Lobato
- Dpto. Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, C/Prof. García González n °2, 41012, Seville, Spain.
| | - Josefa Álvarez-Fuentes
- Dpto. Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, C/Prof. García González n °2, 41012, Seville, Spain
| | - Mercedes Fernández-Arévalo
- Dpto. Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, C/Prof. García González n °2, 41012, Seville, Spain
| | - Lucía Martín-Banderas
- Dpto. Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, C/Prof. García González n °2, 41012, Seville, Spain
| |
Collapse
|
14
|
Prichard KL, O'Brien NS, Murcia SR, Baker JR, McCluskey A. Role of Clathrin and Dynamin in Clathrin Mediated Endocytosis/Synaptic Vesicle Recycling and Implications in Neurological Diseases. Front Cell Neurosci 2022; 15:754110. [PMID: 35115907 PMCID: PMC8805674 DOI: 10.3389/fncel.2021.754110] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
Endocytosis is a process essential to the health and well-being of cell. It is required for the internalisation and sorting of “cargo”—the macromolecules, proteins, receptors and lipids of cell signalling. Clathrin mediated endocytosis (CME) is one of the key processes required for cellular well-being and signalling pathway activation. CME is key role to the recycling of synaptic vesicles [synaptic vesicle recycling (SVR)] in the brain, it is pivotal to signalling across synapses enabling intracellular communication in the sensory and nervous systems. In this review we provide an overview of the general process of CME with a particular focus on two key proteins: clathrin and dynamin that have a central role to play in ensuing successful completion of CME. We examine these two proteins as they are the two endocytotic proteins for which small molecule inhibitors, often of known mechanism of action, have been identified. Inhibition of CME offers the potential to develop therapeutic interventions into conditions involving defects in CME. This review will discuss the roles and the current scope of inhibitors of clathrin and dynamin, providing an insight into how further developments could affect neurological disease treatments.
Collapse
|
15
|
Deluco B, Wilson HL. Assessment of intestinal macromolecular absorption in young piglets to pave the way to oral vaccination: preliminary results. Vet Res Commun 2021; 46:79-91. [PMID: 34559380 PMCID: PMC8461397 DOI: 10.1007/s11259-021-09831-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 09/10/2021] [Indexed: 11/04/2022]
Abstract
The small intestine of the piglet has evolved to be permeable immediately after birth to facilitate the uptake of colostrum-derived immunoglobulins as well as other macromolecules, and cells. However, the precise timing of gut closure in today’s precocious pig is not known. We gavaged piglets immediately after birth and at 1-h after birth with Cy5-labeled Ovalbumin (Cy5-Ova) then harvested their small intestine’s 6–7 h later. To assess localization of Cy5-Ova in the small intestinal epithelial cells, we performed immunohistochemistry using a basolateral surface marker and a recycling endosome marker called pIgR, the late endosomal marker Rab7, and the lysosomal marker LAMP-1. Cy5-Ova co-localized with Rab7 and LAMP-1 in the duodenum and jejunum of 0-h old and 1-h old gavaged piglets, but only in the ileum of 0-h gavaged piglets. These data suggest that movement of Cy5-Ova through the late endosomes to the lysosomes was much reduced in the ileum of 1-h gavaged piglets. Cy5-Ova was largely present in epithelial cell digestive and transport vacuoles, but it did not colocalize with pIgR-positive endosomes in 0-h and 1-h gavaged piglets. Differences in macromolecular uptake across the different regions of the small intestine after only 1-h may be due to prior processing of colostral macromolecules, changes in the intestine due to initiation of colonization by microflora and/or the initiation of gut-closure. Understanding the relationship between the localization of Cy5-Ova and small intestinal permeability may contribute to establishing whether oral vaccination in the newborn can capitalize on the transient permeability before gut closure to promote immune protection.
Collapse
Affiliation(s)
- Brodie Deluco
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada
| | - Heather L Wilson
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada.
| |
Collapse
|
16
|
Chen C, Huang FW, Huang SS, Huang JS. IGFBP-3 and TGF-β inhibit growth in epithelial cells by stimulating type V TGF-β receptor (TβR-V)-mediated tumor suppressor signaling. FASEB Bioadv 2021; 3:709-729. [PMID: 34485840 PMCID: PMC8409558 DOI: 10.1096/fba.2021-00016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/06/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
The TGF-β type V receptor (TβR-V) mediates growth inhibition by IGFBP-3 and TGF-β in epithelial cells and loss of TβR-V expression in these cells leads to development of carcinoma. The mechanisms by which TβR-V mediates growth inhibition (tumor suppressor) signaling remain elusive. Previous studies revealed that IGFBP-3 and TGF-β inhibit growth in epithelial cells by stimulating TβR-V-mediated IRS-1/2-dependent activation and cytoplasm-to-nucleus translocation of IGFBP-3- or TGF-β-stimulated protein phosphatase (PPase), resulting in dephosphorylation of pRb-related proteins (p107, p130) or pRb, and growth arrest. To define the signaling, we characterized/identified the IGFBP-3- and TGF-β-stimulated PPases in cell lysates and nucleus fractions in Mv1Lu cells treated with IGFBP-3 and TGF-β, using a cell-free assay with 32P-labeled casein as a substrate. Both IGFBP-3- and TGF-β-stimulated PPase activities in cell lysates are abolished when cells are co-treated with TGF-β/IGFBP-3 antagonist or RAP (LRP-1/TβR-V antagonist). However, the IGFBP-3-stimulated PPase activity, but not TGF-β-stimulated PPase activity, is sensitive to inhibition by okadaic acid (OA). In addition, OA or PP2Ac siRNA reverses IGFBP-3 growth inhibition, but not TGF-β growth inhibition, in Mv1Lu and 32D cells. These suggest that IGFBP-3- and TGF-β-stimulated PPases are identical to PP2A and PP1, respectively. By Western blot/phosphorimager/immunofluorescence-microscopy analyses, IGFBP-3 and TGF-β stimulate TβR-V-mediated IRS-2-dependent activation and cytoplasm-to-nucleus translocation of PP2Ac and PP1c, resulting in dephosphorylation of p130/p107 and pRb, respectively, and growth arrest. Small molecule TGF-β enhancers, which potentiate TGF-β growth inhibition by enhancing TβR-I-TβR-II-mediated canonical signaling and thus activating TβR-V-mediated tumor suppressor signaling cascade (TβR-V/IRS-2/PP1/pRb), could be used to prevent and treat carcinoma.
Collapse
Affiliation(s)
- Chun‐Lin Chen
- Department of Biological ScienceNational Sun Yat‐sen UniversityKaohsiungTaiwan
- Departments of Biochemistry and Molecular BiologySaint Louis University School of MedicineSt. LouisMOUSA
| | - Franklin W. Huang
- Division of Hematology and OncologyDepartment of MedicineUniversity of CaliforniaSan FranciscoCAUSA
| | | | - Jung San Huang
- Departments of Biochemistry and Molecular BiologySaint Louis University School of MedicineSt. LouisMOUSA
| |
Collapse
|
17
|
Sousa de Almeida M, Susnik E, Drasler B, Taladriz-Blanco P, Petri-Fink A, Rothen-Rutishauser B. Understanding nanoparticle endocytosis to improve targeting strategies in nanomedicine. Chem Soc Rev 2021; 50:5397-5434. [PMID: 33666625 PMCID: PMC8111542 DOI: 10.1039/d0cs01127d] [Citation(s) in RCA: 484] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Indexed: 12/19/2022]
Abstract
Nanoparticles (NPs) have attracted considerable attention in various fields, such as cosmetics, the food industry, material design, and nanomedicine. In particular, the fast-moving field of nanomedicine takes advantage of features of NPs for the detection and treatment of different types of cancer, fibrosis, inflammation, arthritis as well as neurodegenerative and gastrointestinal diseases. To this end, a detailed understanding of the NP uptake mechanisms by cells and intracellular localization is essential for safe and efficient therapeutic applications. In the first part of this review, we describe the several endocytic pathways involved in the internalization of NPs and we discuss the impact of the physicochemical properties of NPs on this process. In addition, the potential challenges of using various inhibitors, endocytic markers and genetic approaches to study endocytosis are addressed along with the principal (semi) quantification methods of NP uptake. The second part focuses on synthetic and bio-inspired substances, which can stimulate or decrease the cellular uptake of NPs. This approach could be interesting in nanomedicine where a high accumulation of drugs in the target cells is desirable and clearance by immune cells is to be avoided. This review contributes to an improved understanding of NP endocytic pathways and reveals potential substances, which can be used in nanomedicine to improve NP delivery.
Collapse
Affiliation(s)
- Mauro Sousa de Almeida
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
| | - Eva Susnik
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
| | - Barbara Drasler
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
| | | | - Alke Petri-Fink
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
- Department of Chemistry, University of FribourgChemin du Musée 91700 FribourgSwitzerland
| | | |
Collapse
|
18
|
Kim B, Lee YE, Yeon JW, Go GY, Byun J, Lee K, Lee HK, Hur JK, Jang M, Kim TH. A novel therapeutic modality using CRISPR-engineered dendritic cells to treat allergies. Biomaterials 2021; 273:120798. [PMID: 33895493 DOI: 10.1016/j.biomaterials.2021.120798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/22/2022]
Abstract
Despite the important roles of dendritic cells (DCs) in airway allergies, current therapeutic strategies such as drugs, allergen immunotherapy and biologics haven't been targeted at them. In this study, we established a promising DC-based therapeutic approach for the alleviation of allergic rhinitis (AR)-associated allergic reactions, using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-mediated targeted gene disruption. RNA sequencing analysis revealed upregulation of vacuolar protein sorting 37 B (VPS37B) in AR-derived DCs, indicating a novel molecular target. Following antigen presentation, VPS37A and VPS37B enabled endocytosis of the mannose receptor, which recognizes the house dust mite (HDM) allergen Der p 1. DCs with targeted disruption of VPS37A/B alleviated Th2 cytokine production when co-cultured in vitro with allogeneic naïve CD4+ T cell from patients with AR. Furthermore, nasal administration of Vps37a/b-disrupted bone marrow DCs to a mouse model of AR resulted in strongly reduced AR-related symptoms. Thus, this novel modality using genetically engineered DCs can provide an effective therapeutic and preventative strategy for allergic diseases.
Collapse
Affiliation(s)
- Byoungjae Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, 02841, Republic of Korea; Neuroscience Research Institute, Korea University, College of Medicine, Seoul, 02841, Republic of Korea
| | - Young Eun Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul, 02792, Republic of Korea; Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Ji Woo Yeon
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Ga-Yeon Go
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul, 02792, Republic of Korea
| | - Junhyoung Byun
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Kijeong Lee
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Hyomin K Lee
- Department of Medicine, Major in Medical Genetics, Graduate School, Hanyang University, Seoul, 04763, Republic of Korea
| | - Junho K Hur
- Department of Genetics, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea; Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Mihue Jang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul, 02792, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Tae Hoon Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
19
|
Rennick JJ, Johnston APR, Parton RG. Key principles and methods for studying the endocytosis of biological and nanoparticle therapeutics. NATURE NANOTECHNOLOGY 2021; 16:266-276. [PMID: 33712737 DOI: 10.1038/s41565-021-00858-8] [Citation(s) in RCA: 704] [Impact Index Per Article: 176.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 01/19/2021] [Indexed: 05/20/2023]
Abstract
Endocytosis is a critical step in the process by which many therapeutic nanomedicines reach their intracellular targets. Our understanding of cellular uptake mechanisms has developed substantially in the past five years. However, these advances in cell biology have not fully translated to the nanoscience and therapeutics literature. Misconceptions surrounding the role of different endocytic pathways and how to study these pathways are hindering progress in developing improved nanoparticle therapies. Here, we summarize the latest insights into cellular uptake mechanisms and pathways. We highlight limitations of current systems to study endocytosis, particularly problems with non-specific inhibitors. We also summarize alternative genetic approaches to robustly probe these pathways and discuss the need to understand how cells endocytose particles in vivo. We hope that this critical assessment of the current methods used in studying nanoparticle uptake will guide future studies at the interface of cell biology and nanomedicine.
Collapse
Affiliation(s)
- Joshua J Rennick
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Brisbane, Queensland, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Brisbane, Queensland, Australia.
| | - Robert G Parton
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Brisbane, Queensland, Australia.
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
20
|
Sheth V, Wang L, Bhattacharya R, Mukherjee P, Wilhelm S. Strategies for Delivering Nanoparticles across Tumor Blood Vessels. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007363. [PMID: 37197212 PMCID: PMC10187772 DOI: 10.1002/adfm.202007363] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Indexed: 05/19/2023]
Abstract
Nanoparticle transport across tumor blood vessels is a key step in nanoparticle delivery to solid tumors. However, the specific pathways and mechanisms of this nanoparticle delivery process are not fully understood. Here, the biological and physical characteristics of the tumor vasculature and the tumor microenvironment are explored and how these features affect nanoparticle transport across tumor blood vessels is discussed. The biological and physical methods to deliver nanoparticles into tumors are reviewed and paracellular and transcellular nanoparticle transport pathways are explored. Understanding the underlying pathways and mechanisms of nanoparticle tumor delivery will inform the engineering of safer and more effective nanomedicines for clinical translation.
Collapse
Affiliation(s)
- Vinit Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, 173 Felgar St, Norman, OK 73019, USA
| | - Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, 173 Felgar St, Norman, OK 73019, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, 800 NE 10th St, Oklahoma City, OK 73104, USA
| | - Priyabrata Mukherjee
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, 800 NE 10th St, Oklahoma City, OK 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, 173 Felgar St, Norman, OK 73019, USA
| |
Collapse
|
21
|
Lownik JC, Conrad DH, Martin RK. T cell receptor signaling defines the fate and pathway of ICOS internalization. Biochem Biophys Rep 2020; 24:100803. [PMID: 32984557 PMCID: PMC7494666 DOI: 10.1016/j.bbrep.2020.100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 10/29/2022] Open
Abstract
The role of the inducible costimulatory of T cells (ICOS) has been shown to be important for many different T cell outcomes and is indispensable for follicular helper T cell (TFH) responses. Since its discovery, there have been several studies on the regulation of ICOS at a transcriptional level. However, the post-translational regulation of ICOS has not been well characterized. Here, we demonstrate that ICOS is internalized following ligation. We show that costimulation with CD3 results in differential internalization and fate than stimulation of ICOS alone. Additionally, we show that ICOS internalization is PI3K and clathrin mediated. The studies presented here not only increase the mechanistic understanding of ICOS post-translational regulation but also give insight into the potential mechanisms by which T cells expressing high affinity receptors may be preferentially chosen to become TFH cells with increased ICOS levels.
Collapse
Affiliation(s)
- Joseph C Lownik
- Center for Clinical and Translational Research, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - Daniel H Conrad
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - Rebecca K Martin
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| |
Collapse
|
22
|
Kim CS, Mathew AP, Uthaman S, Moon MJ, Bae EH, Kim SW, Park IK. Glycol chitosan-based renal docking biopolymeric nanomicelles for site-specific delivery of the immunosuppressant. Carbohydr Polym 2020; 241:116255. [DOI: 10.1016/j.carbpol.2020.116255] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 03/25/2020] [Accepted: 04/02/2020] [Indexed: 01/26/2023]
|
23
|
Soll M, Chen QC, Zhitomirsky B, Lim PP, Termini J, Gray HB, Assaraf YG, Gross Z. Protein-coated corrole nanoparticles for the treatment of prostate cancer cells. Cell Death Discov 2020; 6:67. [PMID: 32793397 PMCID: PMC7387447 DOI: 10.1038/s41420-020-0288-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 02/01/2023] Open
Abstract
Development of novel therapeutic strategies to eradicate malignant tumors is of paramount importance in cancer research. In a recent study, we have introduced a facile protocol for the preparation of corrole-protein nanoparticles (NPs). These NPs consist of a corrole-core coated with protein. We now report that a novel lipophilic corrole, (2)Ga, delivered as human serum albumin (HSA)-coated NPs, displayed antineoplastic activity towards human prostate cancer DU-145 cells. Cryo-TEM analysis of these NPs revealed an average diameter of 50.2 ± 8.1 nm with a spherical architecture exhibiting low polydispersity. In vitro cellular uptake of (2)Ga/albumin NPs was attributable to rapid internalization of the corrole through ligand binding-dependent extracellular release and intercalation of the corrole cargo into the lipid bilayer of the plasma membrane. This finding is in contrast with a previously reported study on corrole-protein NPs that displayed cellular uptake via endocytosis. Investigation of the non-light-induced mechanism of action of (2)Ga suggested the induction of necrosis through plasma membrane destabilization, impairment of calcium homeostasis, lysosomal stress and rupture, as well as formation of reactive oxygen species (ROS). (2)Ga also exhibited potent light-induced cytotoxicity through ROS generation. These findings demonstrate a rapid cellular uptake of (2)Ga/protein NPs along with targeted induction of tumor cell necrosis.
Collapse
Affiliation(s)
- Matan Soll
- Schulich Faculty of Chemistry, Technion–Israel Institute of Technology, 3200003 Haifa, Israel
| | - Qiu-Cheng Chen
- Schulich Faculty of Chemistry, Technion–Israel Institute of Technology, 3200003 Haifa, Israel
| | - Benny Zhitomirsky
- Department of Biology, The Fred Wyszkowski Cancer Research Laboratory, Technion–Israel Institute of Technology, 3200003 Haifa, Israel
| | - Punnajit P. Lim
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Monrovia, CA 91010 USA
| | - John Termini
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Monrovia, CA 91010 USA
| | - Harry B. Gray
- Beckman Institute, California Institute of Technology, Pasadena, CA 91125 USA
| | - Yehuda G. Assaraf
- Department of Biology, The Fred Wyszkowski Cancer Research Laboratory, Technion–Israel Institute of Technology, 3200003 Haifa, Israel
| | - Zeev Gross
- Schulich Faculty of Chemistry, Technion–Israel Institute of Technology, 3200003 Haifa, Israel
| |
Collapse
|
24
|
Kaźmierczak Z, Szostak-Paluch K, Przybyło M, Langner M, Witkiewicz W, Jędruchniewicz N, Dąbrowska K. Endocytosis in cellular uptake of drug delivery vectors: Molecular aspects in drug development. Bioorg Med Chem 2020; 28:115556. [PMID: 32828419 DOI: 10.1016/j.bmc.2020.115556] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/16/2022]
Abstract
Drug delivery vectors are widely applied to increase drug efficacy while reducing the side effects and potential toxicity of a drug. They allow for patient-tailored therapy, dose titration, and therapeutic drug monitoring. A major part of drug delivery systems makes use of large nanocarriers: liposomes or virus-like particles (VLPs). These systems allow for a relatively large amount of cargo with good stability of vectors, and they offer multiple options for targeting vectors in vivo. Here we discuss endocytic pathways that are available for drug delivery by large nanocarriers. We focus on molecular aspects of the process, including an overview of potential molecular targets for studies of drug delivery vectors and for future solutions allowing targeted drug delivery.
Collapse
Affiliation(s)
- Zuzanna Kaźmierczak
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Kamila Szostak-Paluch
- Research and Development Center, Regional Specialized Hospital, Wrocław, Poland; Wrocław University of Science and Technology, Faculty of Fundamental Technical Problems, Department of Biomedical Engineering, Wrocław, Poland
| | - Magdalena Przybyło
- Wrocław University of Science and Technology, Faculty of Fundamental Technical Problems, Department of Biomedical Engineering, Wrocław, Poland; Lipid Systems sp z o.o., Wrocław, Poland
| | - Marek Langner
- Wrocław University of Science and Technology, Faculty of Fundamental Technical Problems, Department of Biomedical Engineering, Wrocław, Poland; Lipid Systems sp z o.o., Wrocław, Poland
| | - Wojciech Witkiewicz
- Research and Development Center, Regional Specialized Hospital, Wrocław, Poland
| | | | - Krystyna Dąbrowska
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland; Research and Development Center, Regional Specialized Hospital, Wrocław, Poland.
| |
Collapse
|
25
|
Martins-Marques T, Catarino S, Gonçalves A, Miranda-Silva D, Gonçalves L, Antunes P, Coutinho G, Leite Moreira A, Falcão Pires I, Girão H. EHD1 Modulates Cx43 Gap Junction Remodeling Associated With Cardiac Diseases. Circ Res 2020; 126:e97-e113. [PMID: 32138615 DOI: 10.1161/circresaha.119.316502] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RATIONALE Efficient communication between heart cells is vital to ensure the anisotropic propagation of electrical impulses, a function mainly accomplished by gap junctions (GJ) composed of Cx43 (connexin 43). Although the molecular mechanisms remain unclear, altered distribution and function of gap junctions have been associated with acute myocardial infarction and heart failure. OBJECTIVE A recent proteomic study from our laboratory identified EHD1 (Eps15 [endocytic adaptor epidermal growth factor receptor substrate 15] homology domain-containing protein 1) as a novel interactor of Cx43 in the heart. METHODS AND RESULTS In the present work, we demonstrate that knockdown of EHD1 impaired the internalization of Cx43, preserving gap junction-intercellular coupling in cardiomyocytes. Interaction of Cx43 with EHD1 was mediated by Eps15 and promoted by phosphorylation and ubiquitination of Cx43. Overexpression of wild-type EHD1 accelerated internalization of Cx43 and exacerbated ischemia-induced lateralization of Cx43 in isolated adult cardiomyocytes. In addition, we show that EHDs associate with Cx43 in human and murine failing hearts. CONCLUSIONS Overall, we identified EHDs as novel regulators of endocytic trafficking of Cx43, participating in the pathological remodeling of gap junctions, paving the way to innovative therapeutic strategies aiming at preserving intercellular communication in the heart.
Collapse
Affiliation(s)
- Tania Martins-Marques
- From the Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (T.M.-M., S.C., L.C., P.A., G.C., H.G.), University of Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (T.M.-M., S.C., H.G.), University of Coimbra, Portugal.,Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.)
| | - Steve Catarino
- From the Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (T.M.-M., S.C., L.C., P.A., G.C., H.G.), University of Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (T.M.-M., S.C., H.G.), University of Coimbra, Portugal.,Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.)
| | - Alexandre Gonçalves
- Department of Surgery and Physiology & Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Portugal (A.G., D.M.S., A.L.M., I.F.P.)
| | - Daniela Miranda-Silva
- Department of Surgery and Physiology & Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Portugal (A.G., D.M.S., A.L.M., I.F.P.)
| | - Lino Gonçalves
- Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.)
| | - Pedro Antunes
- From the Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (T.M.-M., S.C., L.C., P.A., G.C., H.G.), University of Coimbra, Portugal.,Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.).,Cardiothoracic Surgery (P.A., G.C.), Coimbra Hospital and University Centre, Portugal
| | - Gonçalo Coutinho
- From the Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (T.M.-M., S.C., L.C., P.A., G.C., H.G.), University of Coimbra, Portugal.,Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.).,Cardiothoracic Surgery (P.A., G.C.), Coimbra Hospital and University Centre, Portugal
| | - Adelino Leite Moreira
- Department of Surgery and Physiology & Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Portugal (A.G., D.M.S., A.L.M., I.F.P.)
| | - Inês Falcão Pires
- Department of Surgery and Physiology & Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Portugal (A.G., D.M.S., A.L.M., I.F.P.)
| | - Henrique Girão
- From the Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (T.M.-M., S.C., L.C., P.A., G.C., H.G.), University of Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (T.M.-M., S.C., H.G.), University of Coimbra, Portugal.,Clinical Academic Centre of Coimbra, CACC, Portugal (T.M-M., S.C., L.G., P.A., G.C., H.G.)
| |
Collapse
|
26
|
Lübow C, Bockstiegel J, Weindl G. Lysosomotropic drugs enhance pro-inflammatory responses to IL-1β in macrophages by inhibiting internalization of the IL-1 receptor. Biochem Pharmacol 2020; 175:113864. [PMID: 32088265 DOI: 10.1016/j.bcp.2020.113864] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/18/2020] [Indexed: 12/20/2022]
Abstract
Interleukin (IL)-1 signaling leads to production of pro-inflammatory mediators and is regulated by receptor endocytosis. Lysosomotropic drugs have been linked to increased pro-inflammatory responses under sterile inflammatory conditions but the underlying mechanisms have not been fully elucidated. Here, we report that lysosomotropic drugs potentiate pro-inflammatory effects in response to IL-1β via a mechanism involving reactive oxygen species, p38 mitogen-activated protein kinase and reduced IL-1 receptor internalization. Chloroquine and hydroxychloroquine increased IL-1β-induced CXCL8 secretion in macrophages which was critically dependent on the lysosomotropic character and inhibition of macroautophagy but independent from the NLRP3 inflammasome. Co-stimulation with the autophagy inducer interferon gamma attenuated CXCL8 release. Other lysosomotropic drugs like bafilomycin A1, fluoxetine and chlorpromazine but also the endocytosis inhibitor dynasore showed similar pro-inflammatory responses. Increased cell surface expression of IL-1 receptor suggests reduced receptor degradation in the presence of lysosomotropic drugs. Our findings provide new insights into a potentially crucial immunoregulatory mechanism in macrophages that may explain how lysosomotropic drugs drive sterile inflammation.
Collapse
Affiliation(s)
- Charlotte Lübow
- Freie Universität Berlin, Institute of Pharmacy (Pharmacology and Toxicology), Germany; University of Bonn, Pharmaceutical Institute, Section Pharmacology and Toxicology, Germany
| | - Judith Bockstiegel
- University of Bonn, Pharmaceutical Institute, Section Pharmacology and Toxicology, Germany
| | - Günther Weindl
- Freie Universität Berlin, Institute of Pharmacy (Pharmacology and Toxicology), Germany; University of Bonn, Pharmaceutical Institute, Section Pharmacology and Toxicology, Germany.
| |
Collapse
|
27
|
Geng J, Guo X, Wang L, Nguyen RQ, Wang F, Liu C, Wang H. Intracellular Delivery of DNA and Protein by a Novel Cell-Permeable Peptide Derived from DOT1L. Biomolecules 2020; 10:217. [PMID: 32024261 PMCID: PMC7072583 DOI: 10.3390/biom10020217] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/27/2022] Open
Abstract
Cellular uptake and intracellular release efficiency of biomacromolecules is low because of hurdles in the cell membrane that result in limited access to intra-cellular targets with few functional effects. Cell-penetrating peptides (CPPs) act as cargo delivery vehicles to promote therapeutic molecule translocation. Here, we describe the novel CPP-Dot1l that not only penetrates by itself, but also mediates cargo translocation in cultured cells, as confirmed by fluorescence microscopy and fluorescence spectrophotometry. We conducted cytotoxicity assays and safety evaluations, and determined peptide-membrane interactions to understand the possible pathway for cargo translocation. Additional nucleic acid and covalently conjugated green fluorescence protein (GFP) studies mediated by CPP-Dot1l were conducted to show functional delivery potential. Results indicate that CPP-Dot1l is a novel and effective CPP due to its good penetrating properties in different cell lines and its ability to enter cells in a concentration-dependent manner. Its penetration efficiency can be prompted by DMSO pretreatment. In addition, not only can it mediate plasmid delivery, but CPP-Dot1l can also deliver GFP protein into cytosol. In conclusion, the findings of this study showed CPP-Dot1l is an attractive pharmaceutical and biochemical tool for future drug, regenerative medicine, cell therapy, gene therapy, and gene editing-based therapy development.
Collapse
Affiliation(s)
- Jingping Geng
- Department of Pathology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; (J.G.); (X.G.); (L.W.)
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China;
| | - Xiangli Guo
- Department of Pathology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; (J.G.); (X.G.); (L.W.)
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China;
| | - Lidan Wang
- Department of Pathology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; (J.G.); (X.G.); (L.W.)
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China;
| | - Richard Q. Nguyen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Fengqin Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China;
| | - Changbai Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China;
| | - Hu Wang
- Department of Pathology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; (J.G.); (X.G.); (L.W.)
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| |
Collapse
|
28
|
Kumari M, Liu CH, Wu WC. Oligochitosan modified albumin as plasmid DNA delivery vector: Endocytic trafficking, polyplex fate, in vivo compatibility. Int J Biol Macromol 2020; 142:492-502. [DOI: 10.1016/j.ijbiomac.2019.09.121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/15/2019] [Accepted: 09/16/2019] [Indexed: 01/12/2023]
|
29
|
Ayat NR, Sun Z, Sun D, Yin M, Hall RC, Vaidya AM, Liu X, Schilb AL, Scheidt JH, Lu ZR. Formulation of Biocompatible Targeted ECO/siRNA Nanoparticles with Long-Term Stability for Clinical Translation of RNAi. Nucleic Acid Ther 2019; 29:195-207. [PMID: 31140918 PMCID: PMC6686697 DOI: 10.1089/nat.2019.0784] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/29/2019] [Indexed: 12/22/2022] Open
Abstract
Nanoparticle based siRNA formulations often suffer from aggregation and loss of function during storage. We in this study report a frozen targeted RGD-polyethylene glycol (PEG)-ECO/siβ3 nanoparticle formulation with a prolonged shelf life and preserved nanoparticle functionality. The targeted RGD-PEG-ECO/siβ3 nanoparticles are formed by step-wised self-assembly of RGD-PEG-maleimide, ECO, and siRNA. The nanoparticles have a diameter of 224.5 ± 9.41 nm and a zeta potential to 45.96 ± 3.67 mV in water and a size of 234.34 ± 3.01 nm and a near neutral zeta potential in saline solution. The addition of sucrose does not affect their size and zeta potential and substantially preserves the integrity and biological activities of frozen and lyophilized formulations of the targeted nanoparticles. The frozen formulation with as low as 5% sucrose retains nanoparticle integrity (90% siRNA encapsulation), size distribution (polydispersity index [PDI] ≤20%), and functionality (at least 75% silencing efficiency) at -80°C for at least 1 year. The frozen RGD-PEG-ECO/siβ3 nanoparticle formulation exhibits excellent biocompatibility, with no adverse effects on hemocompatibility and minimal immunogenicity. As RNAi holds the promise in treating the previously untreatable diseases, the frozen nanoparticle formulation with the low sucrose concentration has the potential to be a delivery platform for clinical translation of RNAi therapeutics.
Collapse
Affiliation(s)
- Nadia R. Ayat
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Zhanhu Sun
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Da Sun
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Michelle Yin
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Ryan C. Hall
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Amita M. Vaidya
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Xujie Liu
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Andrew L. Schilb
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Josef H. Scheidt
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
30
|
Ju A, Lee SW, Lee YE, Han KC, Kim JC, Shin SC, Park HJ, EunKyeong Kim E, Hong S, Jang M. A carrier-free multiplexed gene editing system applicable for suspension cells. Biomaterials 2019; 217:119298. [PMID: 31280073 DOI: 10.1016/j.biomaterials.2019.119298] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/22/2019] [Indexed: 12/26/2022]
Abstract
Genetically engineered cells via CRISPR/Cas9 system can serve as powerful sources for cancer immunotherapeutic applications. Furthermore, multiple genetic alterations are necessary to overcome tumor-induced immune-suppressive mechanisms. However, one of the major obstacles is the technical difficulty with efficient multiple gene manipulation of suspension cells due to the low transfection efficacy. Herein, we established a carrier-free multiplexed gene editing platform in a simplified method, which can enhance the function of cytotoxic CD8+ T cells by modulating suspension cancer cells. Our multiple Cas9 ribonucleoproteins (RNPs) enable simultaneous disruption of two programmed cell death 1 (PD-1) ligands, functioning as negative regulators in the immune system, by accessing engineered Cas9 proteins with abilities of complexation and cellular penetration. In addition, combination with electroporation enhanced multiple gene editing efficacy, compared with that by treatment of multiple Cas9 RNPs alone. This procedure resulted in high gene editing at multiple loci of suspension cells. The treatment of multiple Cas9 RNPs targeting both ligands strongly improved Th1-type cytokine production of cytotoxic CD8+ T cells, resulting in synergistic cytotoxic effects against cancer. Simultaneous suppression of PD-L1 and PD-L2 on cancer cells via our developed editing system allows effective anti-tumor immunity. Furthermore, the treatment of multiple Cas9 RNPs targeting PD-L1, PD-L2, and TIM-3 had approximately 70-90% deletion efficacy. Thus, our multiplexed gene editing strategy endows potential clinical utilities in cancer immunotherapy.
Collapse
Affiliation(s)
- Anna Ju
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sung Won Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, 05006, Republic of Korea
| | - Young Eun Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 136-701, Republic of Korea
| | - Ki-Cheol Han
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jin-Chul Kim
- Natural Constituents of Research Center, Natural Products Research Institute, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
| | - Sang Chul Shin
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, 05006, Republic of Korea
| | - Eunice EunKyeong Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, 05006, Republic of Korea.
| | - Mihue Jang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
31
|
Ebrahimabadi S, Shahbazi M, Akbari M, Golalipour M, Farazmandfar T. Design and construction of a recombinant lentiviral vector with specific tropism to human epidermal growth factor-overexpressed cancer cells: Developing a new retargeting system for lentivirus vectors. J Gene Med 2019; 21:e3095. [PMID: 31050357 DOI: 10.1002/jgm.3095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Targeting of specific tissues and cells by viruses is one of the challenges faced by researchers. Lentiviral vectors (LVs) are one of the most promising gene delivery systems in cancer gene therapy. Therefore, we aimed to design a novel lentiviral delivery system that expresses anti- human epidermal growth factor 2 (HER2) designed anykrin repeat protein (DARPin) on the vector envelope to create a pseudotyped lentivirus for targeting HER2-positive cancer cells. METHODS A helper plasmid producing the viral vector envelope containing anti-HER2 DARPin-G3 was constructed. LV was produced by transfer vector containing green fluorescent protein (GFP) gene and helper plasmids in human embryonic kidney 293 cells. The human breast cancer cell lines SKBR3 (normal and with inhibited endocytosis) (HER2-positive) and MDA-MB-231 (HER2-negative) were transduced by the recombinant viral vector. The GFP-based transduction rate was determined by flow cytometry and fluorescence microscopy. RESULTS The anti-HER2 DARPin concentration in DARPin-LVs was significantly higher than the envelope G glycoprotein of the vesicular stomatitis virus-LVs (non-anti-HER2 control) (p < 0.0001). In flow cytometry assays, the percentage of transduction by recombinant LV was significantly higher in SKBR3 cells than in SKBR3 cells with inhibited endocytosis (p = 0.0074) and MDA-MB-231 cells (p = 0.0037). In fluorescence microscopy assays, the percentage of transduction by new LV was significantly higher in SKBR3 cells than in SKBR3 cells with inhibited endocytosis (p = 0.0026) and MDA-MB-231 cells (p = 0.0014). CONCLUSIONS We constructed a new recombinant LV with a defect in cell entry directly, containing an anti-HER2 DARPin on the vector envelope with specific tropism to HER2 receptor on HER2-positive cancer cells. We assumed that this viral vector transduces cells via an endocytosis-dependent process.
Collapse
Affiliation(s)
- Sima Ebrahimabadi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mona Akbari
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Masoud Golalipour
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Touraj Farazmandfar
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
32
|
Xiong F, Ling X, Chen X, Chen J, Tan J, Cao W, Ge L, Ma M, Wu J. Pursuing Specific Chemotherapy of Orthotopic Breast Cancer with Lung Metastasis from Docking Nanoparticles Driven by Bioinspired Exosomes. NANO LETTERS 2019; 19:3256-3266. [PMID: 30965009 DOI: 10.1021/acs.nanolett.9b00824] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Breast cancer develops from local tissue but is characterized by a distinct metastatic pattern involving regional lymph nodes and distant organs, which is the primary cause of high mortality in breast cancer patients. Herein, optimal docking nanoparticles (NPs) composed of a laurate-functionalized Pt(IV) prodrug (Pt(lau)), human serum albumin (HSA), and lecithin were predicted by computational modeling, prepared by nanoprecipitation, and validated by fluorescence spectroscopy. As macrophages have been reported to be preferentially recruited by breast cancer, Rex, the exosome spontaneously secreted by murine RAW 264.7 cells, was isolated to encapsulate the NPs. This high-performance delivery system, called NPs/Rex, possessed the desired physicochemical properties, enhanced colloidal stability, and redox-triggered release profile. Investigations of cytodynamics proved that NPs/Rex was internalized through multiple pathways, avoided entrapment by bilayers, and successfully platinized nucleic acids after bioreduction in the cytosol. Intracellular activation of Pt(lau) was confirmed by observing the characteristic effects of cisplatin on cell proliferation and the cell cycle following treatment with NPs/Rex. During in vivo application, the bioinspired Rex coating endowed docking NPs with prolonged blood circulation, smart organ tropism, and enhanced biocompatibility, as well as robust platinum (Pt) chemotherapy for breast cancer cells in orthotopic tumors of fat pads and metastatic nodules of lungs. Therefore, this favorable nanoplatform might provide valuable insight into the derivatization and development of Pt anticancer drugs used currently in the clinic.
Collapse
Affiliation(s)
- Fei Xiong
- School of Biomedical Engineering, State Key Laboratory of Oncology in South China , Sun Yat-sen University , Guangzhou , Guangdong 510006 , China
| | - Xiang Ling
- School of Biomedical Engineering, State Key Laboratory of Oncology in South China , Sun Yat-sen University , Guangzhou , Guangdong 510006 , China
| | - Xing Chen
- School of Biomedical Engineering, State Key Laboratory of Oncology in South China , Sun Yat-sen University , Guangzhou , Guangdong 510006 , China
| | - Jing Chen
- Department of Biological and Environmental Engineering , Cornell University , Ithaca , New York 14853 , United States
| | - Jiaxing Tan
- Life Sciences Institute and Innovation Center for Cell Signaling Network , Zhejiang University , Hangzhou , Zhejiang 310058 , China
| | - Wuji Cao
- Wyss Institute for Biologically Inspired Engineering at Harvard University , Boston , Massachusetts 02115 , United States
| | - Liang Ge
- Department of Pharmaceutics, School of Pharmacy, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , Jiangsu 210009 , China
| | - Minglin Ma
- Department of Biological and Environmental Engineering , Cornell University , Ithaca , New York 14853 , United States
| | - Jun Wu
- School of Biomedical Engineering, State Key Laboratory of Oncology in South China , Sun Yat-sen University , Guangzhou , Guangdong 510006 , China
| |
Collapse
|
33
|
Müller WEG, Ackermann M, Tolba E, Neufurth M, Ivetac I, Kokkinopoulou M, Schröder HC, Wang X. Role of ATP during the initiation of microvascularization: acceleration of an autocrine sensing mechanism facilitating chemotaxis by inorganic polyphosphate. Biochem J 2018; 475:3255-3273. [PMID: 30242064 DOI: 10.1042/bcj20180535] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 12/21/2022]
Abstract
The in vitro tube formation assay with human umbilical vein endothelial cells (HUVEC) was applied to identify the extra- and intracellular sources of metabolic energy/ATP required for cell migration during the initial stage of microvascularization. Extracellularly, the physiological energy-rich polymer, inorganic polyphosphate (polyP), applied as biomimetic amorphous calcium polyP microparticles (Ca-polyP-MP), is functioning as a substrate for ATP generation most likely via the combined action of the alkaline phosphatase (ALP) and the adenylate kinase (AK). The linear Ca-polyP-MP with a size of 40 phosphate units, close to the polyP in the acidocalcisomes in the blood platelets, were found to increase endothelial cell tube formation, as well as the intracellular ATP levels. Depletion of extracellular ATP with apyrase suppressed tube formation during the initial incubation period. Inhibition experiments revealed that inhibitors (levamisole and Ap5A) of the enzymes involved in extracellular ATP generation strongly reduce the Ca-polyP-MP-induced tube formation. The stimulatory effect of Ca-polyP-MP was also diminished by the glycolysis inhibitor oxamate and trifluoperazine which blocks endocytosis, as well as by MRS2211, an antagonist of the P2Y13 receptor. Oligomycin, an inhibitor of the mitochondrial F0F1-ATP synthase, displayed no effect at lower concentrations on tube formation. Electron microscopic data revealed that after cellular uptake, the Ca-polyP-MP accumulate close to the cell membrane. We conclude that in HUVEC exposed to polyP, ATP is formed extracellularly via the coupled ALP-AK reaction, and intracellularly during glycolysis. The results suggest an autocrine signaling pathway of ATP with polyP as an extracellular store of metabolic energy for endothelial cell migration during the initial vascularization process.
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, 55128 Mainz, Germany
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg University, Johann Joachim Becher Weg 13, 55099 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, 55128 Mainz, Germany
- Polymers and Pigments Department, National Research Center, 33 El Buhouth St, Dokki, 12311 Cairo, Egypt
| | - Meik Neufurth
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, Germany
| | - Ivan Ivetac
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, Germany
- Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3002, Australia
| | - Maria Kokkinopoulou
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - 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, 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, 55128 Mainz, Germany
| |
Collapse
|
34
|
Shapira KE, Ehrlich M, Henis YI. Cholesterol depletion enhances TGF-β Smad signaling by increasing c-Jun expression through a PKR-dependent mechanism. Mol Biol Cell 2018; 29:2494-2507. [PMID: 30091670 PMCID: PMC6233055 DOI: 10.1091/mbc.e18-03-0175] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/16/2018] [Accepted: 07/25/2018] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) plays critical roles in numerous physiological and pathological responses. Cholesterol, a major plasma membrane component, can have pronounced effects on signaling responses. Cells continually monitor cholesterol content and activate multilayered transcriptional and translational signaling programs, following perturbations to cholesterol homeostasis (e.g., statins, the commonly used cholesterol-reducing drugs). However, the cross-talk of such programs with ligand-induced signaling responses (e.g., TGF-β signaling) remained unknown. Here, we studied the effects of a mild reduction in free (membrane-associated) cholesterol on distinct components of TGF-β-signaling pathways. Our findings reveal a new regulatory mechanism that enhances TGF-β-signaling responses by acting downstream from receptor activation. Reduced cholesterol results in PKR-dependent eIF2α phosphorylation, which enhances c-Jun translation, leading in turn to higher levels of JNK-mediated c-Jun phosphorylation. Activated c-Jun enhances transcription and expression of Smad2/3. This leads to enhanced sensitivity to TGF-β stimulation, due to increased Smad2/3 expression and phosphorylation. The phospho/total Smad2/3 ratio remains unchanged, indicating that the effect is not due to altered receptor activity. We propose that cholesterol depletion induces overactivation of PKR, JNK, and TGF-β signaling, which together may contribute to the side effects of statins in diverse disease settings.
Collapse
Affiliation(s)
- Keren E. Shapira
- 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
| | - Yoav I. Henis
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
35
|
TOM1 Regulates Neuronal Accumulation of Amyloid-β Oligomers by FcγRIIb2 Variant in Alzheimer's Disease. J Neurosci 2018; 38:9001-9018. [PMID: 30185465 DOI: 10.1523/jneurosci.1996-17.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/16/2018] [Accepted: 08/24/2018] [Indexed: 01/06/2023] Open
Abstract
Emerging evidences suggest that intraneuronal Aβ correlates with the onset of Alzheimer's disease (AD) and highly contributes to neurodegeneration. However, critical mediator responsible for Aβ uptake in AD pathology needs to be clarified. Here, we report that FcγRIIb2, a variant of Fcγ-receptor IIb (FcγRIIb), functions in neuronal uptake of pathogenic Aβ. Cellular accumulation of oligomeric Aβ1-42, not monomeric Aβ1-42 or oligomeric Aβ1-40, was blocked by Fcgr2b knock-out in neurons and partially in astrocytes. Aβ1-42 internalization was FcγRIIb2 di-leucine motif-dependent and attenuated by TOM1, a FcγRIIb2-binding protein that repressed the receptor recycling. TOM1 expression was downregulated in the hippocampus of male 3xTg-AD mice and AD patients, and regulated by miR-126-3p in neuronal cells after exposure to Aβ1-42 In addition, memory impairments in male 3xTg-AD mice were rescued by the lentiviral administration of TOM1 gene. Augmented Aβ uptake into lysosome caused its accumulation in cytoplasm and mitochondria. Moreover, neuronal accumulation of Aβ in both sexes of 3xTg-AD mice and memory deficits in male 3xTg-AD mice were ameliorated by forebrain-specific expression of Aβ-uptake-defective Fcgr2b mutant. Our findings suggest that FcγRIIb2 is essential for neuropathic uptake of Aβ in AD.SIGNIFICANCE STATEMENT Accumulating evidences suggest that intraneuronal Aβ is found in the early step of AD brain and is implicated in the pathogenesis of AD. However, the critical mediator involved in these processes is uncertain. Here, we describe that the FcγRIIb2 variant is responsible for both neuronal uptake and intraneuronal distribution of pathogenic Aβ linked to memory deficits in AD mice, showing a pathologic significance of the internalized Aβ. Further, Aβ internalization is attenuated by TOM1, a novel FcγRIIb2-binding protein. Together, we provide a molecular mechanism responsible for neuronal uptake of pathogenic Aβ found in AD.
Collapse
|
36
|
Protein moiety in oligochitosan modified vector regulates internalization mechanism and gene delivery: Polyplex characterization, intracellular trafficking and transfection. Carbohydr Polym 2018; 202:143-156. [PMID: 30286987 DOI: 10.1016/j.carbpol.2018.08.131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/13/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
Oligochitosan-modified proteins have gained attention as efficient non-viral vectors for gene delivery. However, little information exists if protein moieties can serve as an important role for internalization and endosome escape ability of the genetic material. To explore this issue, we designed two cationic oligochitosan-modified vectors that consist of different proteins, namely a hydrophobic plant protein (zein) and a hydrophilic animal protein (ovalbumin (OVA)) to deliver pDNA to epithelial cell line CHO-K1 and HEK 293 T. These cationic vectors were systematically characterized by molecular weight, infrared (IR) structural analysis, transmission electron microscopy (TEM) morphology, and surface charge. A remarkable impact of protein moieties was observed on physiochemical properties of the developed vectors. Oligochitosan-modified zein containing hydrophobic protein exhibited high buffering capacity and excellent DNA binding ability compared to the oligochitosan-modified OVA. The data on transfection in the presence of endocytic inhibitors indicated that the caveolae-mediated pathway (CvME) played a key role in the internalization of the zein-based polyplex. However, the OVA-based polyplex was internalized in CHO-K1 cells via CvME and in HEK 293 T cells via the lipid-mediated pathway. Moreover, oligochitosan-modified zein exhibited lower cytotoxicity, greater lysosomal escape ability, better plasmid stability, and better transfection efficiency than the oligochitosan-modified OVA. This study offers a facile procedure for the synthesis of cationic vectors and elucidates the relationship that exists between protein moieties and transfection activity, thus providing an alternative, non-viral platform for the gene delivery.
Collapse
|
37
|
Barreau K, Montero-Menei C, Eyer J. The neurofilament derived-peptide NFL-TBS.40-63 enters in-vitro in human neural stem cells and increases their differentiation. PLoS One 2018; 13:e0201578. [PMID: 30092042 PMCID: PMC6084907 DOI: 10.1371/journal.pone.0201578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/18/2018] [Indexed: 12/13/2022] Open
Abstract
Regenerative medicine is a promising approach to treat neurodegenerative diseases by replacing degenerating cells like neurons or oligodendrocytes. Targeting human neural stem cells directly in the brain is a big challenge in such a strategy. The neurofilament derived NFL-TBS.40-63 peptide has recently been introduced as a novel tool to target neural stem cells. Previous studies showed that this peptide can be internalized by rat neural stem cells in vitro and in vivo, which coincided with lower proliferation and self-renewal capacity and increase of differentiation. In this study, we analyzed the uptake and potential effects of the NFL-TBS.40-63 peptide on human neural stem cells isolated from human fetuses. We showed that the peptide inhibits proliferation and the ability to produce neurospheres in vitro, which is consistent with an increase in cell adhesion and differentiation. These results confirm that the peptide could be a promising molecule to target and manipulate human neural stem cells and thus could serve as a strategic tool for regenerative medicine.
Collapse
Affiliation(s)
- Kristell Barreau
- Laboratoire Micro et Nanomédecines Translationnelles, Inserm 1066, CNRS 6021, Institut de Recherche en Ingénierie de la Santé, Bâtiment IBS Institut de Biologie de la Santé, Université Angers, Centre Hospitalier Universitaire, Angers, France
| | - Claudia Montero-Menei
- Centre de Recherche en Cancérologie et Immunologie, INSERM, Université de Nantes, Université Angers, Angers, France
| | - Joël Eyer
- Laboratoire Micro et Nanomédecines Translationnelles, Inserm 1066, CNRS 6021, Institut de Recherche en Ingénierie de la Santé, Bâtiment IBS Institut de Biologie de la Santé, Université Angers, Centre Hospitalier Universitaire, Angers, France
- * E-mail:
| |
Collapse
|
38
|
Radaic A, de Jesus MB. Solid lipid nanoparticles release DNA upon endosomal acidification in human embryonic kidney cells. NANOTECHNOLOGY 2018; 29:315102. [PMID: 29756603 DOI: 10.1088/1361-6528/aac447] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Nanotechnology can produce materials with unique features compared to their bulk counterparts, which can be useful for medical applications (i.e. nanomedicine). Among the therapeutic agents used in nanomedicine, small molecules or biomacromolecules, such as proteins or genetic materials, can be designed for disease diagnostics and treatment. To transport these biomacromolecules to the target cells, nanomedicine requires nanocarriers. Solid lipid nanoparticles (SLNs) are among the promising nanocarriers available, because they can be made from biocompatible materials and present high stability (over one year). In addition, upon the binding genetic material, SLNs form SLNplexes. However, little is yet known about how cells process these SLNplexes-in particular, how internalization and endosome acidification affects the transfection mediated by SLNplexes. Therefore, we aim to investigate how these processes affect SLNplex transfection in HEK293T cells. We find that the SLNplex is mainly internalized by clathrin-mediated endocytosis, which is a fast and reliable pathway to transfection, leading to approximately 60% transfection efficiency. Interestingly, upon acidification (below pH 5.0), the SLN seems to release its DNA content, which can be an essential step for SLNplex transfection. The underlying mechanisms described in this work may help improve SLNplex formulations and transfection efficiency. Moreover, these advances can improve the field of nanomedical research and bring new ways to cure diseases.
Collapse
Affiliation(s)
- A Radaic
- Nano-Cell Interactions Lab., Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | |
Collapse
|
39
|
Soft-shelled turtle iridovirus enters cells via cholesterol-dependent, clathrin-mediated endocytosis as well as macropinocytosis. Arch Virol 2018; 163:3023-3033. [PMID: 30066272 PMCID: PMC7087192 DOI: 10.1007/s00705-018-3966-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 06/03/2018] [Indexed: 12/01/2022]
Abstract
Ranaviruses are nucleoplasmic large DNA viruses that can cause major economic losses in the aquaculture industry and pose a severe threat to global ecological diversity. The available literature demonstrates that classifiable members of the genus Ranavirus enter cells via multiple and complicated routes. Here, we demonstrated the underlying cellular entry mechanism of soft-shelled turtle iridovirus (STIV) using green fluorescence tagged recombinant virus. Treatment with chlorpromazine, sucrose, ethyl-isopropyl amiloride, chloroquine or bafilomycin A1 all significantly decreased STIV infection, suggesting that STIV uses clathrin-mediated endocytosis and macropinocytosis to enter cells via a pH-dependent pathway. Depletion of cellular cholesterol with methyl-β-cyclodextrin significantly inhibited STIV entry, but neither filipin III nor nystatin did, suggesting that STIV entry was cholesterol dependent but caveola independent. Treatment with dynasore, genistein, ML-7 or cytochalasin D all significantly inhibited STIV infection, indicating that Rac GTPase and myosin II activity were required for the macropinocytosis-like pathway as well as actin polymerization. Our findings suggest that the molecular events involved in STIV entry are not identical to those of other ranavirus isolates. Our results also extend our understanding of the molecular mechanism of iridovirus entry and pathogenesis.
Collapse
|
40
|
Shih-Wei W, Chih-Ling C, Kao YC, Martin R, Knölker HJ, Shiao MS, Chen CL. Pentabromopseudilin: a myosin V inhibitor suppresses TGF-β activity by recruiting the type II TGF-β receptor to lysosomal degradation. J Enzyme Inhib Med Chem 2018; 33:920-935. [PMID: 29768059 PMCID: PMC6009923 DOI: 10.1080/14756366.2018.1465416] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pentabromopseudilin (PBrP) is a marine antibiotic isolated from the marine bacteria Pseudomonas bromoutilis and Alteromonas luteoviolaceus. PBrP exhibits antimicrobial, anti-tumour, and phytotoxic activities. In mammalian cells, PBrP is known to act as a reversible and allosteric inhibitor of myosin Va (MyoVa). In this study, we report that PBrP is a potent inhibitor of transforming growth factor-β (TGF-β) activity. PBrP inhibits TGF-β-stimulated Smad2/3 phosphorylation, plasminogen activator inhibitor-1 (PAI-1) protein production and blocks TGF-β-induced epithelial–mesenchymal transition in epithelial cells. PBrP inhibits TGF-β signalling by reducing the cell-surface expression of type II TGF-β receptor (TβRII) and promotes receptor degradation. Gene silencing approaches suggest that MyoVa plays a crucial role in PBrP-induced TβRII turnover and the subsequent reduction of TGF-β signalling. Because, TGF-β signalling is crucial in the regulation of diverse pathophysiological processes such as tissue fibrosis and cancer development, PBrP should be further explored for its therapeutic role in treating fibrotic diseases and cancer.
Collapse
Affiliation(s)
- Wang Shih-Wei
- a Department of Biological Sciences , National Sun Yat-sen University , Kaohsiung , Taiwan, ROC
| | - Chung Chih-Ling
- a Department of Biological Sciences , National Sun Yat-sen University , Kaohsiung , Taiwan, ROC
| | - Yu-Chen Kao
- a Department of Biological Sciences , National Sun Yat-sen University , Kaohsiung , Taiwan, ROC
| | - René Martin
- b Department of Chemistry , TU Dresden , Dresden , Germany
| | | | - Meng-Shin Shiao
- c Faculty of Medicine Ramathibodi Hospital , Mahidol University , Bangkok , Thailand
| | - Chun-Lin Chen
- a Department of Biological Sciences , National Sun Yat-sen University , Kaohsiung , Taiwan, ROC.,d Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica , Kaohsiung , Taiwan, ROC
| |
Collapse
|
41
|
Targeted Delivery of Cell Penetrating Peptide Virus-like Nanoparticles to Skin Cancer Cells. Sci Rep 2018; 8:8499. [PMID: 29855618 PMCID: PMC5981617 DOI: 10.1038/s41598-018-26749-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/18/2018] [Indexed: 01/19/2023] Open
Abstract
Skin cancer or cutaneous carcinoma, is a pre-eminent global public health problem with no signs of plateauing in its incidence. As the most common treatments for skin cancer, surgical resection inevitably damages a patient’s appearance, and chemotherapy has many side effects. Thus, the main aim of this study was to screen for a cell penetrating peptide (CPP) for the development of a targeting vector for skin cancer. In this study, we identified a CPP with the sequence NRPDSAQFWLHH from a phage displayed peptide library. This CPP targeted the human squamous carcinoma A431 cells through an interaction with the epidermal growth factor receptor (EGFr). Methyl-β-cyclodextrin (MβCD) and chlorpromazine hydrochloride (CPZ) inhibited the internalisation of the CPP into the A431 cells, suggesting the peptide entered the cells via clathrin-dependent endocytosis. The CPP displayed on hepatitis B virus-like nanoparticles (VLNPs) via the nanoglue successfully delivered the nanoparticles into A431 cells. The present study demonstrated that the novel CPP can serve as a ligand to target and deliver VLNPs into skin cancer cells.
Collapse
|
42
|
Ganbold T, Baigude H. Design of Mannose-Functionalized Curdlan Nanoparticles for Macrophage-Targeted siRNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:14463-14474. [PMID: 29648784 DOI: 10.1021/acsami.8b02073] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
6-Amino-6-deoxy-curdlan is a promising nucleic acid carrier that efficiently delivers plasmid DNA as well as short interfering RNA (siRNA) to various cell lines. The highly reactive C6-NH2 groups of 6-amino-6-deoxy-curdlan prompt conjugation of various side groups including tissue-targeting ligands to enhance cell-type-specific nucleic acid delivery to specific cell lines. Herein, to test the primary-cell-targeting efficiency of the curdlan derivative, we chemically conjugated a macrophage-targeting ligand, mannose, to 6-amino-6-deoxy-curdlan. The resulting curdlan derivative (denoted CMI) readily complexed with siRNA and formed nanoparticles with a diameter of 50-80 nm. The CMI nanoparticles successfully delivered a dye-labeled siRNA to mouse peritoneal macrophages. The delivery efficiency was blocked by mannan, a natural ligand for a macrophage surface mannose receptor (CD206), but not by zymosan, a ligand for the dectin-1 receptor, which is also present on the surface of macrophages. Moreover, CMI nanoparticles were internalized by macrophages only at 37 °C, suggesting that the cellular uptake of CMI nanoparticles was energy-dependent. Furthermore, CMI nanoparticle efficiently delivered siRNA against tumor necrosis factor α (TNFα) to lipopolysaccharide-stimulated primary mouse peritoneal macrophages. In vivo experiments demonstrated that CMI nanoparticles successfully delivered siTNFα to mouse peritoneal macrophages, liver, and lung and induced significant knockdown of the TNFα expression at both messenger RNA and protein levels. Therefore, our design of CMI may be a promising siRNA carrier for targeting CD206-expressing primary cells such as macrophage and dendritic cells.
Collapse
Affiliation(s)
- Tsogzolmaa Ganbold
- School of Chemistry & Chemical Engineering, Inner Mongolia Key Laboratory of Mongolian Medicinal Chemistry , Inner Mongolia University , 235 West College Road , Hohhot , Inner Mongolia 010020 , P. R. China
| | - Huricha Baigude
- School of Chemistry & Chemical Engineering, Inner Mongolia Key Laboratory of Mongolian Medicinal Chemistry , Inner Mongolia University , 235 West College Road , Hohhot , Inner Mongolia 010020 , P. R. China
| |
Collapse
|
43
|
Chung CL, Wang SW, Sun WC, Shu CW, Kao YC, Shiao MS, Chen CL. Sorafenib suppresses TGF-β responses by inducing caveolae/lipid raft-mediated internalization/degradation of cell-surface type II TGF-β receptors: Implications in development of effective adjunctive therapy for hepatocellular carcinoma. Biochem Pharmacol 2018; 154:39-53. [PMID: 29678520 DOI: 10.1016/j.bcp.2018.04.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/13/2018] [Indexed: 12/31/2022]
Abstract
Sorafenib is the only FDA approved drug for the treatment of advanced hepatocellular carcinoma (HCC) and other malignancies. Studies indicate that TGF-β signalling is associated with tumour progression in HCC. Autocrine and paracrine TGF-β promotes tumour growth and malignancy by inducing epithelial-mesenchymal transition (EMT). Sorafenib is believed to antagonize tumour progression by inhibiting TGF-β-induced EMT. It improves survival of patients but HCC later develops resistance and relapses. The underlying mechanism of resistance is unknown. Understanding of the molecular mechanism of sorafenib inhibition of TGF-β-induced signalling or responses in HCC may lead to development of adjunctive effective therapy for HCC. In this study, we demonstrate that sorafenib suppresses TGF-β responsiveness in hepatoma cells, hepatocytes, and animal liver, mainly by downregulating cell-surface type II TGF-β receptors (TβRII) localized in caveolae/lipid rafts and non-lipid raft microdomains via caveolae/lipid rafts-mediated internalization and degradation. Furthermore, sorafenib-induced downregulation and degradation of cell-surface TβRII is prevented by simultaneous treatment with a caveolae disruptor or lysosomal inhibitors. On the other hand, sorafenib only downregulates cell-surface TβRII localized in caveolae/lipid rafts but not localized in non-lipid raft microdomains in hepatic stellate cells. These results suggest that sorafenib inhibits TGF-β signalling mainly by inducing caveolae/lipid raft-mediated internalization and degradation of cell-surface TβR-II in target cells. They may also imply that treatment with agents which promote formation of caveolae/lipid rafts, TGF-β receptor kinase inhibitors (e.g., LY2157299) or TGF-β peptide antagonists (by liver-targeting delivery) may be considered as effective adjunct therapy with sorafenib for HCC.
Collapse
Affiliation(s)
- Chih-Ling Chung
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC
| | - Shih-Wei Wang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC
| | - Wei-Chih Sun
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81326, Taiwan, ROC
| | - Chih-Wen Shu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81326, Taiwan, ROC
| | - Yu-Chen Kao
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC
| | - Meng-Shin Shiao
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan, ROC.
| |
Collapse
|
44
|
Tassew NG, Charish J, Shabanzadeh AP, Luga V, Harada H, Farhani N, D'Onofrio P, Choi B, Ellabban A, Nickerson PEB, Wallace VA, Koeberle PD, Wrana JL, Monnier PP. Exosomes Mediate Mobilization of Autocrine Wnt10b to Promote Axonal Regeneration in the Injured CNS. Cell Rep 2018; 20:99-111. [PMID: 28683327 DOI: 10.1016/j.celrep.2017.06.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/19/2017] [Accepted: 05/28/2017] [Indexed: 12/29/2022] Open
Abstract
Developing strategies that promote axonal regeneration within the injured CNS is a major therapeutic challenge, as axonal outgrowth is potently inhibited by myelin and the glial scar. Although regeneration can be achieved using the genetic deletion of PTEN, a negative regulator of the mTOR pathway, this requires inactivation prior to nerve injury, thus precluding therapeutic application. Here, we show that, remarkably, fibroblast-derived exosomes (FD exosomes) enable neurite growth on CNS inhibitory proteins. Moreover, we demonstrate that, upon treatment with FD exosomes, Wnt10b is recruited toward lipid rafts and activates mTOR via GSK3β and TSC2. Application of FD exosomes shortly after optic nerve injury promoted robust axonal regeneration, which was strongly reduced in Wnt10b-deleted animals. This work uncovers an intercellular signaling pathway whereby FD exosomes mobilize an autocrine Wnt10b-mTOR pathway, thereby awakening the intrinsic capacity of neurons for regeneration, an important step toward healing the injured CNS.
Collapse
Affiliation(s)
- Nardos G Tassew
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON M5T 2S8, Canada
| | - Jason Charish
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON M5T 2S8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Alireza P Shabanzadeh
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON M5T 2S8, Canada
| | - Valbona Luga
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, 982 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Hidekiyo Harada
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON M5T 2S8, Canada
| | - Nahal Farhani
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON M5T 2S8, Canada
| | - Philippe D'Onofrio
- Department of Anatomy, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Brian Choi
- Department of Anatomy, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Ahmad Ellabban
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON M5T 2S8, Canada
| | - Philip E B Nickerson
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON M5T 2S8, Canada
| | - Valerie A Wallace
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON M5T 2S8, Canada; Department of Ophthalmology and Vision Science, Faculty of Medicine, University of Toronto, 340 College Street, Toronto, ON M5T 3A9, Canada
| | - Paulo D Koeberle
- Department of Anatomy, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Jeffrey L Wrana
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, 982 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Philippe P Monnier
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON M5T 2S8, Canada; Department of Ophthalmology and Vision Science, Faculty of Medicine, University of Toronto, 340 College Street, Toronto, ON M5T 3A9, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
45
|
Chung CL, Wang SW, Martin R, Knölker HJ, Kao YC, Lin MH, Chen JJ, Huang YB, Wu DC, Chen CL. Pentachloropseudilin Inhibits Transforming Growth Factor-β (TGF-β) Activity by Accelerating Cell-Surface Type II TGF-β Receptor Turnover in Target Cells. Chembiochem 2018; 19:851-864. [DOI: 10.1002/cbic.201700693] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Chih-Ling Chung
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
| | - Shih-Wei Wang
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
| | - René Martin
- Department of Chemistry; Technische Universität Dresden; Bergstrasse 66 01069 Dresden Germany
| | - Hans-Joachim Knölker
- Department of Chemistry; Technische Universität Dresden; Bergstrasse 66 01069 Dresden Germany
| | - Yu-Chen Kao
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
| | - Ming-Hong Lin
- Department of Microbiology and Immunology; Faculty of Medicine; Kaohsiung Medical University Hospital; Kaohsiung 80708 ROC Taiwan
| | - Jih-Jung Chen
- Faculty of Pharmacy; School of Pharmaceutical Sciences; National Yang-Ming University; Taipei 11221 ROC Taiwan
| | - Yaw-Bin Huang
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
- Department of Pharmacy; School of Pharmacy; Kaohsiung Medical University; Kaohsiung 80708 ROC Taiwan
- Center for Stem Cell Research; Kaohsiung Medical University; Kaohsiung 80708 ROC Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology; Department of Internal Medicine; Kaohsiung Medical University Hospital; Kaohsiung 80708 ROC Taiwan
- Center for Stem Cell Research; Kaohsiung Medical University; Kaohsiung 80708 ROC Taiwan
| | - Chun-Lin Chen
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
- Doctoral Degree Program in Marine Biotechnology; National Sun Yat-sen University and Academia Sinica; Kaohsiung 80424 ROC Taiwan
| |
Collapse
|
46
|
Abdelkader DH, Osman MA, El-Gizawy SA, Hawthorne SJ, Faheem AM, McCarron PA. Effect of poly(ethylene glycol) on insulin stability and cutaneous cell proliferation in vitro following cytoplasmic delivery of insulin-loaded nanoparticulate carriers – A potential topical wound management approach. Eur J Pharm Sci 2018; 114:372-384. [PMID: 29288081 DOI: 10.1016/j.ejps.2017.12.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 12/14/2022]
|
47
|
Alshehri A, Grabowska A, Stolnik S. Pathways of cellular internalisation of liposomes delivered siRNA and effects on siRNA engagement with target mRNA and silencing in cancer cells. Sci Rep 2018; 8:3748. [PMID: 29491352 PMCID: PMC5830644 DOI: 10.1038/s41598-018-22166-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/19/2018] [Indexed: 01/01/2023] Open
Abstract
Design of an efficient delivery system is a generally recognised bottleneck in translation of siRNA technology into clinic. Despite research efforts, cellular processes that determine efficiency of siRNA silencing achieved by different delivery formulations remain unclear. Here, we investigated the mechanism(s) of cellular internalisation of a model siRNA-loaded liposome system in a correlation to the engagement of delivered siRNA with its target and consequent silencing by adopting siRNA molecular beacon technology. Probing of cellular internalisation pathways by a panel of pharmacological inhibitors indicated that clathrin-mediated (dynamin-dependent) endocytosis, macropinocytosis (dynamine independent), and cell membrane cholesterol dependent process(es) (clathrin and caveolea-independent) all play a role in the siRNA-liposomes internalization. The inhibition of either of these entry routes was, in general, mirrored by a reduction in the level of siRNA engagement with its target mRNA, as well as in a reduction of the target gene silencing. A dramatic increase in siRNA engagement with its target RNA was observed on disruption of endosomal membrane (by chloroquine), accompanied with an increased silencing. The work thus illustrates that employing molecular beacon siRNA technology one can start to assess the target RNA engagement - a stage between initial cellular internalization and final gene silencing of siRNA delivery systems.
Collapse
Affiliation(s)
- Abdullah Alshehri
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Anna Grabowska
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Snow Stolnik
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
48
|
Ferguson R, Subramanian V. The cellular uptake of angiogenin, an angiogenic and neurotrophic factor is through multiple pathways and largely dynamin independent. PLoS One 2018; 13:e0193302. [PMID: 29486010 PMCID: PMC5828446 DOI: 10.1371/journal.pone.0193302] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 02/08/2018] [Indexed: 01/25/2023] Open
Abstract
Angiogenin (ANG), a member of the RNase superfamily (also known as RNase 5) has neurotrophic, neuroprotective and angiogenic activities. Recently it has also been shown to be important in stem cell homeostasis. Mutations in ANG are associated with neurodegenerative diseases such as Amyotrophic Lateral Sclerosis (ALS) and Fronto-temporal dementia (FTD). ANG is a secreted protein which is taken up by cells and translocated to the nucleus. However, the import pathway/s through which ANG is taken up is/are still largely unclear. We have characterised the uptake of ANG in neuronal, astrocytic and microglial cell lines as well as primary neurons and astrocytes using pharmacological agents as well as dominant negative dynamin and Rab5 to perturb uptake and intracellular trafficking. We find that uptake of ANG is largely clathrin/dynamin independent and microtubule depolymerisation has a marginal effect. Perturbation of membrane ruffling and macropinocytosis significantly inhibited ANG uptake suggesting an uptake mechanism similar to RNase A. Our findings shed light on why mutations which do not overtly affect RNase activity but cause impaired localization are associated with neurodegenerative disease.
Collapse
Affiliation(s)
- Ross Ferguson
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Vasanta Subramanian
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
- * E-mail:
| |
Collapse
|
49
|
Müller WEG, Wang S, Wiens M, Neufurth M, Ackermann M, Relkovic D, Kokkinopoulou M, Feng Q, Schröder HC, Wang X. Uptake of polyphosphate microparticles in vitro (SaOS-2 and HUVEC cells) followed by an increase of the intracellular ATP pool size. PLoS One 2017; 12:e0188977. [PMID: 29287071 PMCID: PMC5747424 DOI: 10.1371/journal.pone.0188977] [Citation(s) in RCA: 23] [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: 09/07/2017] [Accepted: 11/16/2017] [Indexed: 12/19/2022] Open
Abstract
Recently two approaches were reported that addressed a vitally important problem in regenerative medicine, i. e. the successful treatment of wounds even under diabetic conditions. Accordingly, these studies with diabetic rabbits [Sarojini et al. PLoS One 2017, 12(4):e0174899] and diabetic mice [Müller et al. Polymers 2017, 9, 300] identified a novel (potential) target for the acceleration of wound healing in diabetes. Both studies propose a raise of the intracellular metabolic energy status via exogenous administration either of ATP, encapsulated into lipid vesicles, or of polyphosphate (polyP) micro-/nanoparticles. Recently this physiological polymer, polyP, was found to release metabolic energy in form of ATP into both the extra- and also intra-cellular space. In the present work the uptake mechanism of the amorphous polyP microparticles "Ca-polyP-MP" has been described and found to be a clathrin-dependent endocytosis import, based on inhibition studies with the inhibitor trifluoperazine, which blocks the clathrin-dependent endocytosis import. The experiments had been performed with SaOS-2 cells, by studying the uptake and distribution of the electron-dense particles into the cells, and with HUVEC cells, for analysis of the intracellular accumulation of polyP, visualized by fluorescent staining of polyP. Concurrently with the uptake of particular polyP the intracellular ATP level increased as well. In contrast to "Ca-polyP-MP" the soluble polyP, administered as "Na-polyP[Ca2+]", did not cause an increase in the intracellular Ca2+ level, suggesting a different mode of action of these two forms of polyP. Based on existing data on the effect of polyP and ATP on the induction of vascularization during wound repair, both groups (Sarojini et al. and Müller et al.) propose that the acceleration of wound repair is based on an increased metabolic energy supply directly to the regenerating wound area.
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, Mainz, Duesbergweg 6, Mainz, Germany
| | - Shunfeng Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Duesbergweg 6, Mainz, Germany
| | - Matthias Wiens
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Duesbergweg 6, Mainz, Germany
| | - Meik Neufurth
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Duesbergweg 6, Mainz, Germany
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Dinko Relkovic
- Fidelta Ltd., Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Maria Kokkinopoulou
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, Germany; and
| | - Qingling Feng
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, China
| | - Heinz C. Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Duesbergweg 6, Mainz, Germany
| | - Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Duesbergweg 6, Mainz, Germany
| |
Collapse
|
50
|
Yuan D, Zhao Y, Banks WA, Bullock KM, Haney M, Batrakova E, Kabanov AV. Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain. Biomaterials 2017; 142:1-12. [PMID: 28715655 DOI: 10.1016/j.biomaterials.2017.07.011] [Citation(s) in RCA: 440] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/04/2017] [Accepted: 07/09/2017] [Indexed: 12/17/2022]
Abstract
Recent work has stimulated interest in the use of exosomes as nanocarriers for delivery of small drugs, RNAs, and proteins to the central nervous system (CNS). To overcome the blood-brain barrier (BBB), exosomes were modified with brain homing peptides that target brain endothelium but likely to increase immune response. Here for the first time we demonstrate that there is no need for such modification to penetrate the BBB in mammals. The naïve macrophage (Mϕ) exosomes can utilize, 1) on the one hand, the integrin lymphocyte function-associated antigen 1 (LFA-1) and intercellular adhesion molecule 1 (ICAM-1), and, 2) on the other hand, the carbohydrate-binding C-type lectin receptors, to interact with brain microvessel endothelial cells comprising the BBB. Notably, upregulation of ICAM-1, a common process in inflammation, promotes Mϕ exosomes uptake in the BBB cells. We further demonstrate in vivo that naïve Mϕ exosomes, after intravenous (IV) administration, cross the BBB and deliver a cargo protein, the brain derived neurotrophic factor (BDNF), to the brain. This delivery is enhanced in the presence of brain inflammation, a condition often present in CNS diseases. Taken together, the findings are of interest to basic science and possible use of Mϕ-derived exosomes as nanocarriers for brain delivery of therapeutic proteins to treat CNS diseases.
Collapse
Affiliation(s)
- Dongfen Yuan
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Yuling Zhao
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - William A Banks
- Research and Development Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Kristin M Bullock
- Research and Development Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
| | - Matthew Haney
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Elena Batrakova
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, USA; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia.
| |
Collapse
|