1
|
Henke L, Ghorbani A, Mole SE. The use of nanocarriers in treating Batten disease: A systematic review. Int J Pharm 2025; 670:125094. [PMID: 39694161 DOI: 10.1016/j.ijpharm.2024.125094] [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: 09/12/2024] [Revised: 12/09/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
The neuronal ceroid lipofuscinoses, commonly known as Batten disease, are a group of lysosomal storage disorders affecting children. There is extensive central nervous system and retinal degeneration, resulting in seizures, vision loss and a progressive cognitive and motor decline. Enzyme replacement and gene therapies are being developed, and mRNA and oligonucleotide therapies are more recently being considered. Overcoming the challenges of the blood-brain barrier and blood-ocular barrier is crucial for effectively targeting the brain and eye, whatever the therapeutic approach. Nanoparticles and extracellular vesicles are small carriers that can encapsulate a cargo and pass through these cell barriers. They have been investigated as drug carriers for other pathologies and could be a promising treatment strategy for Batten disease. Their use in gene, enzyme, or mRNA replacement therapy of all lysosomal storage disorders, including Mucopolysaccharidoses, Niemann-Pick diseases, and Fabry disease, is investigated in this systematic review. Different nanocarriers can efficiently target the lysosome and cross the barriers into the brain and eyes. This supports continued exploration of nanocarriers as potential future treatment options for Batten disease.
Collapse
Affiliation(s)
- Larissa Henke
- Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Ali Ghorbani
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Sara E Mole
- Great Ormond Street Institute of Child Health, University College London, London WC1E 6BT, UK.
| |
Collapse
|
2
|
Zamora ME, Essien EO, Bhamidipati K, Murthy A, Liu J, Kim H, Patel MN, Nong J, Wang Z, Espy C, Chaudhry FN, Ferguson LT, Tiwari S, Hood ED, Marcos-Contreras OA, Omo-Lamai S, Shuvaeva T, Arguiri E, Wu J, Rauova L, Poncz M, Basil MC, Cantu E, Planer JD, Spiller K, Zepp J, Muzykantov VR, Myerson JW, Brenner JS. Marginated Neutrophils in the Lungs Effectively Compete for Nanoparticles Targeted to the Endothelium, Serving as a Part of the Reticuloendothelial System. ACS NANO 2024; 18:22275-22297. [PMID: 39105696 DOI: 10.1021/acsnano.4c06286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Nanomedicine has long pursued the goal of targeted delivery to specific organs and cell types but has yet to achieve this goal with the vast majority of targets. One rare example of success in this pursuit has been the 25+ years of studies targeting the lung endothelium using nanoparticles conjugated to antibodies against endothelial surface molecules. However, here we show that such "endothelial-targeted" nanocarriers also effectively target the lungs' numerous marginated neutrophils, which reside in the pulmonary capillaries and patrol for pathogens. We show that marginated neutrophils' uptake of many of these "endothelial-targeted" nanocarriers is on par with endothelial uptake. This generalizes across diverse nanomaterials and targeting moieties and was even found with physicochemical lung tropism (i.e., without targeting moieties). Further, we observed this in ex vivo human lungs and in vivo healthy mice, with an increase in marginated neutrophil uptake of nanoparticles caused by local or distant inflammation. These findings have implications for nanomedicine development for lung diseases. These data also suggest that marginated neutrophils, especially in the lungs, should be considered a major part of the reticuloendothelial system (RES), with a special role in clearing nanoparticles that adhere to the lumenal surfaces of blood vessels.
Collapse
Affiliation(s)
- Marco E Zamora
- Drexel University School of Biomedical Engineering, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Eno-Obong Essien
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Kartik Bhamidipati
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Aditi Murthy
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Jing Liu
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Hyunjun Kim
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Manthan N Patel
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jia Nong
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Zhicheng Wang
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Carolann Espy
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Fatima N Chaudhry
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Laura T Ferguson
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Sachchidanand Tiwari
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Elizabeth D Hood
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A Marcos-Contreras
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Serena Omo-Lamai
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Tea Shuvaeva
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Evguenia Arguiri
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jichuan Wu
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Lubica Rauova
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Mortimer Poncz
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Maria C Basil
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Edward Cantu
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Joseph D Planer
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Kara Spiller
- Drexel University School of Biomedical Engineering, Philadelphia, Pennsylvania 19104, United States
| | - Jarod Zepp
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R Muzykantov
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jacob W Myerson
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jacob S Brenner
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
3
|
Pan Y, Chen J, Zhang Y, Ren Y, Wu Z, Xue Q, Zeng S, Fang C, Zhang H, Zhang L, Liu C, Zeng J. Second Near-Infrared Macrophage-Biomimetic Nanoprobes for Photoacoustic Imaging of Neuroinflammation. Mol Pharm 2024; 21:1804-1816. [PMID: 38466359 DOI: 10.1021/acs.molpharmaceut.3c01115] [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] [Indexed: 03/13/2024]
Abstract
Neuroinflammation is a significant pathological event involving the neurodegenerative process associated with many neurological disorders. Diagnosis and treatment of neuroinflammation in its early stage are essential for the prevention and management of neurological diseases. Herein, we designed macrophage membrane-coated photoacoustic (PA) probes (MSINPs), with targeting specificities based on naturally existing target-ligand interactions for the early diagnosis of neuroinflammation. The second near-infrared dye, IR1061, was doped into silica as the core and was encapsulated with a macrophage membrane. In vitro as well as in vivo, the MSINPs could target inflammatory cells via the inflammation chemotactic effect. PA imaging was used to trace the MSINPs in a neuroinflammation mouse model and showed a great targeted effect of MSINPs in the prefrontal cortex. Therefore, the biomimetic nanoprobe prepared in this study offers a new strategy for PA molecular imaging of neuroinflammation, which can enhance our understanding of the evolution of neuroinflammation in specific brain regions.
Collapse
Affiliation(s)
- Yingying Pan
- Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Jingqin Chen
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuling Zhang
- Shenzhen Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518116, China
| | - Yaguang Ren
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhifeng Wu
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qiang Xue
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Ultrasound, Shenzhen People's Hospital, The Second Clinical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Silue Zeng
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chihua Fang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Hai Zhang
- Department of Ultrasound, Shenzhen People's Hospital, The Second Clinical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Lingyan Zhang
- Lab of Molecular Imaging and Medical Intelligence, Department of Radiology, Longgang Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Chengbo Liu
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jie Zeng
- Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| |
Collapse
|
4
|
Loeck M, Placci M, Muro S. Effect of acid sphingomyelinase deficiency in type A Niemann-Pick disease on the transport of therapeutic nanocarriers across the blood-brain barrier. Drug Deliv Transl Res 2023; 13:3077-3093. [PMID: 37341882 DOI: 10.1007/s13346-023-01374-z] [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] [Accepted: 05/20/2023] [Indexed: 06/22/2023]
Abstract
ASM deficiency in Niemann-Pick disease type A results in aberrant cellular accumulation of sphingomyelin, neuroinflammation, neurodegeneration, and early death. There is no available treatment because enzyme replacement therapy cannot surmount the blood-brain barrier (BBB). Nanocarriers (NCs) targeted across the BBB via transcytosis might help; yet, whether ASM deficiency alters transcytosis remains poorly characterized. We investigated this using model NCs targeted to intracellular adhesion molecule-1 (ICAM-1), transferrin receptor (TfR), or plasmalemma vesicle-associated protein-1 (PV1) in ASM-normal vs. ASM-deficient BBB models. Disease differentially changed the expression of all three targets, with ICAM-1 becoming the highest. Apical binding and uptake of anti-TfR NCs and anti-PV1 NCs were unaffected by disease, while anti-ICAM-1 NCs had increased apical binding and decreased uptake rate, resulting in unchanged intracellular NCs. Additionally, anti-ICAM-1 NCs underwent basolateral reuptake after transcytosis, whose rate was decreased by disease, as for apical uptake. Consequently, disease increased the effective transcytosis rate for anti-ICAM-1 NCs. Increased transcytosis was also observed for anti-PV1 NCs, while anti-TfR NCs remained unaffected. A fraction of each formulation trafficked to endothelial lysosomes. This was decreased in disease for anti-ICAM-1 NCs and anti-PV1 NCs, agreeing with opposite transcytosis changes, while it increased for anti-TfR NCs. Overall, these variations in receptor expression and NC transport resulted in anti-ICAM-1 NCs displaying the highest absolute transcytosis in the disease condition. Furthermore, these results revealed that ASM deficiency can differently alter these processes depending on the particular target, for which this type of study is key to guide the design of therapeutic NCs.
Collapse
Affiliation(s)
- Maximilian Loeck
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marina Placci
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain.
- Institution of Catalonia for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
5
|
Lu Q, Liu T, Han Z, Zhao J, Fan X, Wang H, Song J, Ye H, Sun J. Revolutionizing cancer treatment: The power of cell-based drug delivery systems. J Control Release 2023; 361:604-620. [PMID: 37579974 DOI: 10.1016/j.jconrel.2023.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/30/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Intravenous administration of drugs is a widely used cancer therapy approach. However, the efficacy of these drugs is often hindered by various biological barriers, including circulation, accumulation, and penetration, resulting in poor delivery to solid tumors. Recently, cell-based drug delivery platforms have emerged as promising solutions to overcome these limitations. These platforms offer several advantages, including prolonged circulation time, active targeting, controlled release, and excellent biocompatibility. Cell-based delivery systems encompass cell membrane coating, intracellular loading, and extracellular backpacking. These innovative platforms hold the potential to revolutionize cancer diagnosis, monitoring, and treatment, presenting a plethora of opportunities for the advancement and integration of pharmaceuticals, medicine, and materials science. Nevertheless, several technological, ethical, and financial barriers must be addressed to facilitate the translation of these platforms into clinical practice. In this review, we explore the emerging strategies to overcome these challenges, focusing specifically on the functions and advantages of cell-mediated drug delivery in cancer treatment.
Collapse
Affiliation(s)
- Qi Lu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Tian Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Zeyu Han
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jian Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Xiaoyuan Fan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Helin Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jiaxuan Song
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Hao Ye
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China; Multi-Scale Robotics Lab (MSRL), Institute of Robotics & Intelligent Systems (IRIS), ETH Zurich, Zurich 8092, Switzerland.
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
6
|
Okamoto Y, Shikano S. Emerging roles of a chemoattractant receptor GPR15 and ligands in pathophysiology. Front Immunol 2023; 14:1179456. [PMID: 37457732 PMCID: PMC10348422 DOI: 10.3389/fimmu.2023.1179456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Chemokine receptors play a central role in the maintenance of immune homeostasis and development of inflammation by directing leukocyte migration to tissues. GPR15 is a G protein-coupled receptor (GPCR) that was initially known as a co-receptor for human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV), with structural similarity to other members of the chemoattractant receptor family. Since the discovery of its novel function as a colon-homing receptor of T cells in mice a decade ago, GPR15 has been rapidly gaining attention for its involvement in a variety of inflammatory and immune disorders. The recent identification of its natural ligand C10orf99, a chemokine-like polypeptide strongly expressed in gastrointestinal tissues, has established that GPR15-C10orf99 is a novel signaling axis that controls intestinal homeostasis and inflammation through the migration of immune cells. In addition, it has been demonstrated that C10orf99-independent functions of GPR15 and GPR15-independent activities of C10orf99 also play significant roles in the pathophysiology. Therefore, GPR15 and its ligands are potential therapeutic targets. To provide a basis for the future development of GPR15- or GPR15 ligand-targeted therapeutics, we have summarized the latest advances in the role of GPR15 and its ligands in human diseases as well as the molecular mechanisms that regulate GPR15 expression and functions.
Collapse
Affiliation(s)
| | - Sojin Shikano
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
7
|
Placci M, Giannotti MI, Muro S. Polymer-based drug delivery systems under investigation for enzyme replacement and other therapies of lysosomal storage disorders. Adv Drug Deliv Rev 2023; 197:114683. [PMID: 36657645 PMCID: PMC10629597 DOI: 10.1016/j.addr.2022.114683] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/30/2022] [Accepted: 12/25/2022] [Indexed: 01/18/2023]
Abstract
Lysosomes play a central role in cellular homeostasis and alterations in this compartment associate with many diseases. The most studied example is that of lysosomal storage disorders (LSDs), a group of 60 + maladies due to genetic mutations affecting lysosomal components, mostly enzymes. This leads to aberrant intracellular storage of macromolecules, altering normal cell function and causing multiorgan syndromes, often fatal within the first years of life. Several treatment modalities are available for a dozen LSDs, mostly consisting of enzyme replacement therapy (ERT) strategies. Yet, poor biodistribution to main targets such as the central nervous system, musculoskeletal tissue, and others, as well as generation of blocking antibodies and adverse effects hinder effective LSD treatment. Drug delivery systems are being studied to surmount these obstacles, including polymeric constructs and nanoparticles that constitute the focus of this article. We provide an overview of the formulations being tested, the diseases they aim to treat, and the results observed from respective in vitro and in vivo studies. We also discuss the advantages and disadvantages of these strategies, the remaining gaps of knowledge regarding their performance, and important items to consider for their clinical translation. Overall, polymeric nanoconstructs hold considerable promise to advance treatment for LSDs.
Collapse
Affiliation(s)
- Marina Placci
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Marina I Giannotti
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; CIBER-BBN, ISCIII, Barcelona, Spain; Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona 08028, Spain
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; Institute of Catalonia for Research and Advanced Studies (ICREA), Barcelona 08010, Spain; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
8
|
Pfrieger FW. The Niemann-Pick type diseases – A synopsis of inborn errors in sphingolipid and cholesterol metabolism. Prog Lipid Res 2023; 90:101225. [PMID: 37003582 DOI: 10.1016/j.plipres.2023.101225] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Disturbances of lipid homeostasis in cells provoke human diseases. The elucidation of the underlying mechanisms and the development of efficient therapies represent formidable challenges for biomedical research. Exemplary cases are two rare, autosomal recessive, and ultimately fatal lysosomal diseases historically named "Niemann-Pick" honoring the physicians, whose pioneering observations led to their discovery. Acid sphingomyelinase deficiency (ASMD) and Niemann-Pick type C disease (NPCD) are caused by specific variants of the sphingomyelin phosphodiesterase 1 (SMPD1) and NPC intracellular cholesterol transporter 1 (NPC1) or NPC intracellular cholesterol transporter 2 (NPC2) genes that perturb homeostasis of two key membrane components, sphingomyelin and cholesterol, respectively. Patients with severe forms of these diseases present visceral and neurologic symptoms and succumb to premature death. This synopsis traces the tortuous discovery of the Niemann-Pick diseases, highlights important advances with respect to genetic culprits and cellular mechanisms, and exposes efforts to improve diagnosis and to explore new therapeutic approaches.
Collapse
|
9
|
Liu GW, Guzman EB, Menon N, Langer RS. Lipid Nanoparticles for Nucleic Acid Delivery to Endothelial Cells. Pharm Res 2023; 40:3-25. [PMID: 36735106 PMCID: PMC9897626 DOI: 10.1007/s11095-023-03471-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023]
Abstract
Endothelial cells play critical roles in circulatory homeostasis and are also the gateway to the major organs of the body. Dysfunction, injury, and gene expression profiles of these cells can cause, or are caused by, prevalent chronic diseases such as diabetes, cardiovascular disease, and cancer. Modulation of gene expression within endothelial cells could therefore be therapeutically strategic in treating longstanding disease challenges. Lipid nanoparticles (LNP) have emerged as potent, scalable, and tunable carrier systems for delivering nucleic acids, making them attractive vehicles for gene delivery to endothelial cells. Here, we discuss the functions of endothelial cells and highlight some receptors that are upregulated during health and disease. Examples and applications of DNA, mRNA, circRNA, saRNA, siRNA, shRNA, miRNA, and ASO delivery to endothelial cells and their targets are reviewed, as well as LNP composition and morphology, formulation strategies, target proteins, and biomechanical factors that modulate endothelial cell targeting. Finally, we discuss FDA-approved LNPs as well as LNPs that have been tested in clinical trials and their challenges, and provide some perspectives as to how to surmount those challenges.
Collapse
Affiliation(s)
- Gary W Liu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Edward B Guzman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Nandita Menon
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Strand Therapeutics, MA, 02215, Boston, USA
| | - Robert S Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
10
|
Abstract
This Review examines the state-of-the-art in the delivery of nucleic acid therapies that are directed to the vascular endothelium. First, we review the most important homeostatic functions and properties of the vascular endothelium and summarize the nucleic acid tools that are currently available for gene therapy and nucleic acid delivery. Second, we consider the opportunities available with the endothelium as a therapeutic target and the experimental models that exist to evaluate the potential of those opportunities. Finally, we review the progress to date from investigations that are directly targeting the vascular endothelium: for vascular disease, for peri-transplant therapy, for angiogenic therapies, for pulmonary endothelial disease, and for the blood-brain barrier, ending with a summary of the future outlook in this field.
Collapse
Affiliation(s)
| | | | | | - W. Mark Saltzman
- Department of Biomedical Engineering
- Department of Chemical & Environmental Engineering
- Department of Cellular & Molecular Physiology
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06510
| |
Collapse
|
11
|
Chountoulesi M, Perinelli DR, Forys A, Chrysostomou V, Kaminari A, Bonacucina G, Trzebicka B, Pispas S, Demetzos C. Development of stimuli-responsive lyotropic liquid crystalline nanoparticles targeting lysosomes: Physicochemical, morphological and drug release studies. Int J Pharm 2022; 630:122440. [PMID: 36436746 DOI: 10.1016/j.ijpharm.2022.122440] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/10/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
The abilities of sub-cellular targeting and stimuli-responsiveness are critical challenges in pharmaceutical nanotechnology. In the present study, glyceryl monooleate (GMO)-based non-lamellar lyotropic liquid crystalline nanoparticles were stabilized by the poly(2-(dimethylamino)ethyl methacrylate)-b-poly(lauryl methacrylate) block copolymer carrying tri-phenyl-phosphine cations (TPP-QPDMAEMA-b-PLMA), either used alone or in combination with other polymers as co-stabilizers. The systems were designed to perform simultaneously sub-cellular targeting, stimuli-responsiveness and to exhibit stealthiness. The physicochemical characteristics and fractal dimensions of the resultant nanosystems were obtained from light scattering techniques, while their micropolarity and microfluidity from fluorescence spectroscopy. Their morphology was assessed by cryo-TEM, while their thermal behavior by microcalorimetry and high-resolution ultrasound spectroscopy. The analyzed properties, including the responsiveness to pH and temperature, were found to be dependent on the combination of the polymeric stabilizers. The subcellular localization was monitored by confocal microscopy, revealing targeting to lysosomes. Subsequently, resveratrol was loaded into the nanosystems, the entrapment efficiency was investigated and in vitro release studies were carried out at different conditions, in which a stimuli-triggered drug release profile was achieved. In conclusion, the proposed multi-functional nanosystems can be considered as potentially stealth, stimuli-responsive drug delivery nanocarriers, with targeting ability to lysosomes and presenting a stimuli-triggered drug release profile.
Collapse
Affiliation(s)
- Maria Chountoulesi
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou 15771, Athens, Greece
| | - Diego Romano Perinelli
- School of Pharmacy, Chemistry Interdisciplinary Project (CHIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Aleksander Forys
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34 ul. M. Curie-Skłodowskiej, Zabrze, Poland
| | - Varvara Chrysostomou
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou 15771, Athens, Greece; Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Archontia Kaminari
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Giulia Bonacucina
- School of Pharmacy, Chemistry Interdisciplinary Project (CHIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Barbara Trzebicka
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34 ul. M. Curie-Skłodowskiej, Zabrze, Poland
| | - Stergios Pispas
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou 15771, Athens, Greece.
| |
Collapse
|
12
|
Tomsen-Melero J, Merlo-Mas J, Carreño A, Sala S, Córdoba A, Veciana J, González-Mira E, Ventosa N. Liposomal formulations for treating lysosomal storage disorders. Adv Drug Deliv Rev 2022; 190:114531. [PMID: 36089182 DOI: 10.1016/j.addr.2022.114531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/13/2022] [Accepted: 09/04/2022] [Indexed: 01/24/2023]
Abstract
Lysosomal storage disorders (LSD) are a group of rare life-threatening diseases caused by a lysosomal dysfunction, usually due to the lack of a single enzyme required for the metabolism of macromolecules, which leads to a lysosomal accumulation of specific substrates, resulting in severe disease manifestations and early death. There is currently no definitive cure for LSD, and despite the approval of certain therapies, their effectiveness is limited. Therefore, an appropriate nanocarrier could help improve the efficacy of some of these therapies. Liposomes show excellent properties as drug carriers, because they can entrap active therapeutic compounds offering protection, biocompatibility, and selectivity. Here, we discuss the potential of liposomes for LSD treatment and conduct a detailed analysis of promising liposomal formulations still in the preclinical development stage from various perspectives, including treatment strategy, manufacturing, characterization, and future directions for implementing liposomal formulations for LSD.
Collapse
Affiliation(s)
- Judit Tomsen-Melero
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red - Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | | | - Aida Carreño
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red - Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Santi Sala
- Nanomol Technologies SL, 08193 Cerdanyola del Vallès, Spain
| | - Alba Córdoba
- Nanomol Technologies SL, 08193 Cerdanyola del Vallès, Spain
| | - Jaume Veciana
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red - Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Elisabet González-Mira
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red - Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Nora Ventosa
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain; Centro de Investigación Biomédica en Red - Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| |
Collapse
|
13
|
Solomon M, Loeck M, Silva-Abreu M, Moscoso R, Bautista R, Vigo M, Muro S. Altered blood-brain barrier transport of nanotherapeutics in lysosomal storage diseases. J Control Release 2022; 349:1031-1044. [PMID: 35901858 PMCID: PMC10550198 DOI: 10.1016/j.jconrel.2022.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/02/2022] [Accepted: 07/19/2022] [Indexed: 12/24/2022]
Abstract
Treatment of neurological lysosomal storage disorders (LSDs) are limited because of impermeability of the blood-brain barrier (BBB) to macromolecules. Nanoformulations targeting BBB transcytosis are being explored, but the status of these routes in LSDs is unknown. We studied nanocarriers (NCs) targeted to the transferrin receptor (TfR), ganglioside GM1 or ICAM1, associated to the clathrin, caveolar or cell adhesion molecule (CAM) routes, respectively. We used brain endothelial cells and mouse models of acid sphingomyelinase-deficient Niemann Pick disease (NPD), and postmortem LSD patients' brains, all compared to respective controls. NC transcytosis across brain endothelial cells and brain distribution in mice were affected, yet through different mechanisms. Reduced TfR and clathrin expression were found, along with decreased transcytosis in cells and mouse brain distribution. Caveolin-1 expression and GM1 transcytosis were also reduced, yet increased GM1 levels seemed to compensate, providing similar NC brain distribution in NPD vs. control mice. A tendency to lower NHE-1 levels was seen, but highly increased ICAM1 expression in cells and human brains correlated with increased transcytosis and brain distribution in mice. Thus, transcytosis-related alterations in NPD and likely other LSDs may impact therapeutic access to the brain, illustrating the need for these mechanistic studies.
Collapse
Affiliation(s)
- Melani Solomon
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA.
| | - Maximilian Loeck
- Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marcelle Silva-Abreu
- Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Ronaldo Moscoso
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA
| | - Ronelle Bautista
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA
| | - Marco Vigo
- Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA; Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain; Institute of Catalonia for Research and Advanced Studies, Barcelona, Spain.
| |
Collapse
|
14
|
Li YX, Wang HB, Li J, Jin JB, Hu JB, Yang CL. Targeting pulmonary vascular endothelial cells for the treatment of respiratory diseases. Front Pharmacol 2022; 13:983816. [PMID: 36110525 PMCID: PMC9468609 DOI: 10.3389/fphar.2022.983816] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022] Open
Abstract
Pulmonary vascular endothelial cells (VECs) are the main damaged cells in the pathogenesis of various respiratory diseases and they mediate the development and regulation of the diseases. Effective intervention targeting pulmonary VECs is of great significance for the treatment of respiratory diseases. A variety of cell markers are expressed on the surface of VECs, some of which can be specifically combined with the drugs or carriers modified by corresponding ligands such as ICAM-1, PECAM-1, and P-selectin, to achieve effective delivery of drugs in lung tissues. In addition, the great endothelial surface area of the pulmonary vessels, the “first pass effect” of venous blood in lung tissues, and the high volume and relatively slow blood perfusion rate of pulmonary capillaries further promote the drug distribution in lung tissues. This review summarizes the representative markers at the onset of respiratory diseases, drug delivery systems designed to target these markers and their therapeutic effects.
Collapse
Affiliation(s)
- Yi-Xuan Li
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, China
| | - Hong-Bo Wang
- Department of Pharmacy, Yuyao People’s Hospital, Yuyao, China
| | - Jing Li
- Department of Pharmacy, Yuyao People’s Hospital, Yuyao, China
| | - Jian-Bo Jin
- Department of Pharmacy, Yuyao People’s Hospital, Yuyao, China
| | - Jing-Bo Hu
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, China
- *Correspondence: Jing-Bo Hu, ; Chun-Lin Yang,
| | - Chun-Lin Yang
- Department of Pharmacy, Yuyao People’s Hospital, Yuyao, China
- *Correspondence: Jing-Bo Hu, ; Chun-Lin Yang,
| |
Collapse
|
15
|
Muntimadugu E, Silva-Abreu M, Vives G, Loeck M, Pham V, del Moral M, Solomon M, Muro S. Comparison between Nanoparticle Encapsulation and Surface Loading for Lysosomal Enzyme Replacement Therapy. Int J Mol Sci 2022; 23:ijms23074034. [PMID: 35409394 PMCID: PMC8999373 DOI: 10.3390/ijms23074034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 12/27/2022] Open
Abstract
Poly(lactide-co-glycolide) (PLGA) nanoparticles (NPs) enhance the delivery of therapeutic enzymes for replacement therapy of lysosomal storage disorders. Previous studies examined NPs encapsulating or coated with enzymes, but these formulations have never been compared. We examined this using hyaluronidase (HAse), deficient in mucopolysaccharidosis IX, and acid sphingomyelinase (ASM), deficient in types A−B Niemann−Pick disease. Initial screening of size, PDI, ζ potential, and loading resulted in the selection of the Lactel II co-polymer vs. Lactel I or Resomer, and Pluronic F68 surfactant vs. PVA or DMAB. Enzyme input and addition of carrier protein were evaluated, rendering NPs having, e.g., 181 nm diameter, 0.15 PDI, −36 mV ζ potential, and 538 HAse molecules encapsulated per NP. Similar NPs were coated with enzyme, which reduced loading (e.g., 292 HAse molecules/NP). NPs were coated with targeting antibodies (> 122 molecules/NP), lyophilized for storage without alterations, and acceptably stable at physiological conditions. NPs were internalized, trafficked to lysosomes, released active enzyme at lysosomal conditions, and targeted both peripheral organs and the brain after i.v. administration in mice. While both formulations enhanced enzyme delivery compared to free enzyme, encapsulating NPs surpassed coated counterparts (18.4- vs. 4.3-fold enhancement in cells and 6.2- vs. 3-fold enhancement in brains), providing guidance for future applications.
Collapse
Affiliation(s)
- Eameema Muntimadugu
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (E.M.); (V.P.); (M.S.)
| | - Marcelle Silva-Abreu
- Institute for Bioengineering of Catalonia, Barcelona Institute for Science and Technology, 08028 Barcelona, Spain; (M.S.-A.); (G.V.); (M.L.); (M.d.M.)
| | - Guillem Vives
- Institute for Bioengineering of Catalonia, Barcelona Institute for Science and Technology, 08028 Barcelona, Spain; (M.S.-A.); (G.V.); (M.L.); (M.d.M.)
| | - Maximilian Loeck
- Institute for Bioengineering of Catalonia, Barcelona Institute for Science and Technology, 08028 Barcelona, Spain; (M.S.-A.); (G.V.); (M.L.); (M.d.M.)
| | - Vy Pham
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (E.M.); (V.P.); (M.S.)
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA
| | - Maria del Moral
- Institute for Bioengineering of Catalonia, Barcelona Institute for Science and Technology, 08028 Barcelona, Spain; (M.S.-A.); (G.V.); (M.L.); (M.d.M.)
| | - Melani Solomon
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (E.M.); (V.P.); (M.S.)
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (E.M.); (V.P.); (M.S.)
- Institute for Bioengineering of Catalonia, Barcelona Institute for Science and Technology, 08028 Barcelona, Spain; (M.S.-A.); (G.V.); (M.L.); (M.d.M.)
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA
- Catalan Institution for Research and Advanced Studies, 08010 Barcelona, Spain
- Correspondence:
| |
Collapse
|
16
|
Xiao T, He M, Xu F, Fan Y, Jia B, Shen M, Wang H, Shi X. Macrophage Membrane-Camouflaged Responsive Polymer Nanogels Enable Magnetic Resonance Imaging-Guided Chemotherapy/Chemodynamic Therapy of Orthotopic Glioma. ACS NANO 2021; 15:20377-20390. [PMID: 34860014 DOI: 10.1021/acsnano.1c08689] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Development of innovative nanomedicine formulations to traverse the blood-brain barrier (BBB) for effective theranostics of glioma remains a great challenge. Herein, we report the creation of macrophage membrane-camouflaged multifunctional polymer nanogels coloaded with manganese dioxide (MnO2) and cisplatin for magnetic resonance (MR) imaging-guided chemotherapy/chemodynamic therapy (CDT) of orthotopic glioma. Redox-responsive poly(N-vinylcaprolactam) (PVCL) nanogels (NGs) formed via precipitation polymerization were in situ loaded with MnO2 and physically encapsulated with cisplatin to have a mean size of 106.3 nm and coated with macrophage membranes to have a good colloidal stability. The generated hybrid NGs display dual pH- and redox-responsive cisplatin and Mn(II) release profiles and can deplete glutathione (GSH) rich in tumor microenvironment through reaction with disulfide-containing cross-linkers within the NGs and MnO2. The thus created Mn(II) enables enhanced CDT through a Fenton-like reaction and T1-weighted MR imaging, while the loaded cisplatin not only exerts its chemotherapy effect but also promotes the reactive oxygen species generation to enhance the CDT efficacy. Importantly, the macrophage membrane coating rendered the hybrid NGs with prolonged blood circulation time and ability to traverse BBB for specific targeted chemotherapy/CDT of orthotopic glioma. Our study demonstrates a promising self-adaptive and cooperative NG-based nanomedicine platform for highly efficient theranostics of glioma, which may be extended to tackle other difficult cancer types.
Collapse
Affiliation(s)
- Tingting Xiao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People's Republic of China
| | - Meijuan He
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Fang Xu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People's Republic of China
| | - Yu Fan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People's Republic of China
| | - Bingyang Jia
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People's Republic of China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People's Republic of China
| | - Han Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People's Republic of China
| |
Collapse
|
17
|
Qamar B, Solomon M, Marin A, Fuerst TR, Andrianov AK, Muro S. Intracellular Delivery of Active Proteins by Polyphosphazene Polymers. Pharmaceutics 2021; 13:249. [PMID: 33578893 PMCID: PMC7916676 DOI: 10.3390/pharmaceutics13020249] [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: 12/30/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
Achieving intracellular delivery of protein therapeutics within cells remains a significant challenge. Although custom formulations are available for some protein therapeutics, the development of non-toxic delivery systems that can incorporate a variety of active protein cargo and maintain their stability, is a topic of great relevance. This study utilized ionic polyphosphazenes (PZ) that can assemble into supramolecular complexes through non-covalent interactions with different types of protein cargo. We tested a PEGylated graft copolymer (PZ-PEG) and a pyrrolidone containing linear derivative (PZ-PYR) for their ability to intracellularly deliver FITC-avidin, a model protein. In endothelial cells, PZ-PYR/protein exhibited both faster internalization and higher uptake levels than PZ-PEG/protein, while in cancer cells both polymers achieved similar uptake levels over time, although the internalization rate was slower for PZ-PYR/protein. Uptake was mediated by endocytosis through multiple mechanisms, PZ-PEG/avidin colocalized more profusely with endo-lysosomes, and PZ-PYR/avidin achieved greater cytosolic delivery. Consequently, a PZ-PYR-delivered anti-F-actin antibody was able to bind to cytosolic actin filaments without needing cell permeabilization. Similarly, a cell-impermeable Bax-BH3 peptide known to induce apoptosis, decreased cell viability when complexed with PZ-PYR, demonstrating endo-lysosomal escape. These biodegradable PZs were non-toxic to cells and represent a promising platform for drug delivery of protein therapeutics.
Collapse
Affiliation(s)
- Bareera Qamar
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA;
| | - Melani Solomon
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (M.S.); (A.M.); (T.R.F.)
| | - Alexander Marin
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (M.S.); (A.M.); (T.R.F.)
| | - Thomas R. Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (M.S.); (A.M.); (T.R.F.)
| | - Alexander K. Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (M.S.); (A.M.); (T.R.F.)
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (M.S.); (A.M.); (T.R.F.)
- Institute of Catalonia for Research and Advanced Studies, 08010 Barcelona, Spain
- Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| |
Collapse
|
18
|
Edelmann MJ, Maegawa GHB. CNS-Targeting Therapies for Lysosomal Storage Diseases: Current Advances and Challenges. Front Mol Biosci 2020; 7:559804. [PMID: 33304924 PMCID: PMC7693645 DOI: 10.3389/fmolb.2020.559804] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022] Open
Abstract
During the past decades, several therapeutic approaches have been developed and made rapidly available for many patients afflicted with lysosomal storage disorders (LSDs), inborn organelle disorders with broad clinical manifestations secondary to the progressive accumulation of undegraded macromolecules within lysosomes. These conditions are individually rare, but, collectively, their incidence ranges from 1 in 2,315 to 7,700 live-births. Most LSDs are manifested by neurological symptoms or signs, including developmental delay, seizures, acroparesthesia, motor weakness, and extrapyramidal signs. The chronic and later-onset clinical forms are at one end of the continuum spectrum and are characterized by a subtle and slow progression of neurological symptoms. Due to its inherent physiological properties, unfortunately, the blood-brain barrier (BBB) constitutes a significant obstacle for current and upcoming therapies to achieve the central nervous system (CNS) and treat neurological problems so prevalent in these conditions. To circumvent this limitation, several strategies have been developed to make the therapeutic agent achieve the CNS. This narrative will provide an overview of current therapeutic strategies under development to permeate the BBB, and address and unmet need for treatment of the progressive neurological manifestations, which are so prevalent in these inherited lysosomal disorders.
Collapse
Affiliation(s)
- Mariola J Edelmann
- Department of Microbiology and Cell Science, The University of Florida's Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Gustavo H B Maegawa
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
19
|
Manthe RL, Loeck M, Bhowmick T, Solomon M, Muro S. Intertwined mechanisms define transport of anti-ICAM nanocarriers across the endothelium and brain delivery of a therapeutic enzyme. J Control Release 2020; 324:181-193. [PMID: 32389778 PMCID: PMC7720842 DOI: 10.1016/j.jconrel.2020.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
The interaction of drug delivery systems with tissues is key for their application. An example is drug carriers targeted to endothelial barriers, which can be transported to intra-endothelial compartments (lysosomes) or transcellularly released at the tissue side (transcytosis). Although carrier targeting valency influences this process, the mechanism is unknown. We studied this using polymer nanocarriers (NCs) targeted to intercellular adhesion molecule-1 (ICAM-1), an endothelial-surface glycoprotein whose expression is increased in pathologies characterized by inflammation. A bell-shaped relationship was found between NC targeting valency and the rate of transcytosis, where high and low NC valencies rendered less efficient transcytosis rates than an intermediate valency formulation. In contrast, an inverted bell-shape relationship was found for NC valency and lysosomal trafficking rates. Data suggested a model where NC valency plays an opposing role in the two sub-processes involved in transcytosis: NC binding-uptake depended directly on valency and exocytosis-detachment was inversely related to this parameter. This is because the greater the avidity of the NC-receptor interaction the more efficient uptake becomes, but NC-receptor detachment post-transport is more compromised. Cleavage of the receptor at the basolateral side of endothelial cells facilitated NC transcytosis, likely by helping NC detachment post-transport. Since transcytosis encompasses both sets of events, the full process finds an optimum at the intersection of these inverted relationships, explaining the bell-shaped behavior. NCs also trafficked to lysosomes from the apical side and, additionally, from the basolateral side in the case of high valency NCs which are slower at detaching from the receptor. This explains the opposite behavior of NC valency for transcytosis vs. lysosomal transport. Anti-ICAM NCs were verified to traffic into the brain after intravenous injection in mice, and both cellular and in vivo data showed that intermediate valency NCs resulted in higher delivery of a therapeutic enzyme, acid sphingomyelinase, required for types A and B Niemann-Pick disease.
Collapse
Affiliation(s)
- Rachel L Manthe
- Institute for Bioscience and Biotechnology Research (IBBR) and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA
| | - Maximilian Loeck
- Institute for Bioengineering of Catalonia (IBEC) of the Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Tridib Bhowmick
- Institute for Bioscience and Biotechnology Research (IBBR) and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA
| | - Melani Solomon
- Institute for Bioscience and Biotechnology Research (IBBR) and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research (IBBR) and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA; Institute for Bioengineering of Catalonia (IBEC) of the Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain; Institution of Catalonia for Research and Advanced Studies (ICREA), Barcelona 08910, Spain.
| |
Collapse
|
20
|
Kiseleva RY, Glassman PG, LeForte KM, Walsh LR, Villa CH, Shuvaev VV, Myerson JW, Aprelev PA, Marcos-Contreras OA, Muzykantov VR, Greineder CF. Bivalent engagement of endothelial surface antigens is critical to prolonged surface targeting and protein delivery in vivo. FASEB J 2020; 34:11577-11593. [PMID: 32738178 DOI: 10.1096/fj.201902515rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/20/2022]
Abstract
Targeted drug delivery to the endothelium has the potential to generate localized therapeutic effects at the blood-tissue interface. For some therapeutic cargoes, it is essential to maintain contact with the bloodstream to exert protective effects. The pharmacokinetics (PK) of endothelial surface-targeted affinity ligands and biotherapeutic cargo remain a largely unexplored area, despite obvious translational implications for this strategy. To bridge this gap, we site-specifically radiolabeled mono- (scFv) and bivalent (mAb) affinity ligands specific for the endothelial cell adhesion molecules, PECAM-1 (CD31) and ICAM-1 (CD54). Radiotracing revealed similar lung biodistribution at 30 minutes post-injection (79.3% ± 4.2% vs 80.4% ± 10.6% ID/g for αICAM and 58.9% ± 3.6% ID/g vs. 47.7% ± 5.8% ID/g for αPECAM mAb vs. scFv), but marked differences in organ residence time, with antibodies demonstrating an order of magnitude greater area under the lung concentration vs. time curve (AUCinf 1698 ± 352 vs. 53.3 ± 7.9 ID/g*hrs for αICAM and 1023 ± 507 vs. 114 ± 37 ID/g*hrs for αPECAM mAb vs scFv). A physiologically based pharmacokinetic model, fit to and validated using these data, indicated contributions from both superior binding characteristics and prolonged circulation time supporting multiple binding-detachment cycles. We tested the ability of each affinity ligand to deliver a prototypical surface cargo, thrombomodulin (TM), using one-to-one protein conjugates. Bivalent mAb-TM was superior to monovalent scFv-TM in both pulmonary targeting and lung residence time (AUCinf 141 ± 3.2 vs 12.4 ± 4.2 ID/g*hrs for ICAM and 188 ± 90 vs 34.7 ± 19.9 ID/g*hrs for PECAM), despite having similar blood PK, indicating that binding strength is more important parameter than the kinetics of binding. To maximize bivalent target engagement, we synthesized an oriented, end-to-end anti-ICAM mAb-TM conjugate and found that this therapeutic had the best lung residence time (AUCinf 253 ± 18 ID/g*hrs) of all TM modalities. These observations have implications not only for the delivery of TM, but also potentially all therapeutics targeted to the endothelial surface.
Collapse
Affiliation(s)
- R Yu Kiseleva
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - P G Glassman
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - K M LeForte
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - L R Walsh
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - C H Villa
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - V V Shuvaev
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - J W Myerson
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - P A Aprelev
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - O A Marcos-Contreras
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - V R Muzykantov
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - C F Greineder
- Department of Emergency Medicine and Pharmacology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
21
|
Mahmoud AM, Morrow JP, Pizzi D, Azizah AM, Davis TP, Tabor RF, Kempe K. Tuning Cellular Interactions of Carboxylic Acid-Side-Chain-Containing Polyacrylates: The Role of Cyanine Dye Label and Side-Chain Type. Biomacromolecules 2020; 21:3007-3016. [PMID: 32598140 DOI: 10.1021/acs.biomac.0c00244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cellular uptake and intracellular targeting to specific organelles are key events in the cellular processing of nanomaterials. Herein, we perform a detailed structure-property relationship study on carboxylic acid-side-chain-bearing polyacrylates to provide design criteria for the manipulation of their cellular interactions. Redox-initiated reversible addition-fragmentation chain-transfer (RRAFT) polymerization of three tert-butyl-protected N-acylated amino ester-based acrylate monomers of different substitutions and degrees of polymerization (DPs) yielded defined and pH-responsive carboxylic acid-side-chain polymers upon deprotection (N-acetyl, DP 1: P(M1); N-propionyl, DP 1: P(E1), DP 2: P(E2)). Flow cytometry studies revealed time-dependent cell association with P(E2) > P(E1) > P(M1) at any given time point. Importantly, the type of cyanine dye used for labeling was found to significantly influence the cellular processing of the polymers. Changing the dye from Cy5 to its sulfonated version sulfoCy5 resulted in a much lower cellular association. Moreover, Cy5-labeled polymers were targeted to mitochondria, while sulfoCy5 modification caused a significant change in the cellular fate of polymers toward lysosome trafficking. This study highlights the importance of selecting a suitable dye but also demonstrates the possibilities for the rational design of organelle-specific targeting of carboxylated polyacrylates.
Collapse
Affiliation(s)
- Ayaat M Mahmoud
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Joshua P Morrow
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - David Pizzi
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Ailsa M Azizah
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.,Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Rico F Tabor
- School of Chemistry, Monash University, Clayton, VIC 3800, Australia
| | - Kristian Kempe
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.,Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
22
|
Glassman PM, Myerson JW, Ferguson LT, Kiseleva RY, Shuvaev VV, Brenner JS, Muzykantov VR. Targeting drug delivery in the vascular system: Focus on endothelium. Adv Drug Deliv Rev 2020; 157:96-117. [PMID: 32579890 PMCID: PMC7306214 DOI: 10.1016/j.addr.2020.06.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
The bloodstream is the main transporting pathway for drug delivery systems (DDS) from the site of administration to the intended site of action. In many cases, components of the vascular system represent therapeutic targets. Endothelial cells, which line the luminal surface of the vasculature, play a tripartite role of the key target, barrier, or victim of nanomedicines in the bloodstream. Circulating DDS may accumulate in the vascular areas of interest and in off-target areas via mechanisms bypassing specific molecular recognition, but using ligands of specific vascular determinant molecules enables a degree of precision, efficacy, and specificity of delivery unattainable by non-affinity DDS. Three decades of research efforts have focused on specific vascular targeting, which have yielded a multitude of DDS, many of which are currently undergoing a translational phase of development for biomedical applications, including interventions in the cardiovascular, pulmonary, and central nervous systems, regulation of endothelial functions, host defense, and permeation of vascular barriers. We discuss the design of endothelial-targeted nanocarriers, factors underlying their interactions with cells and tissues, and describe examples of their investigational use in models of acute vascular inflammation with an eye on translational challenges.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America.
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Laura T Ferguson
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Raisa Y Kiseleva
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Vladimir V Shuvaev
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America.
| |
Collapse
|
23
|
Hsueh PY, Ju Y, Vega A, Edman MC, MacKay JA, Hamm-Alvarez SF. A Multivalent ICAM-1 Binding Nanoparticle which Inhibits ICAM-1 and LFA-1 Interaction Represents a New Tool for the Investigation of Autoimmune-Mediated Dry Eye. Int J Mol Sci 2020; 21:ijms21082758. [PMID: 32326657 PMCID: PMC7216292 DOI: 10.3390/ijms21082758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/11/2020] [Accepted: 04/12/2020] [Indexed: 12/31/2022] Open
Abstract
The autoimmune disorder, Sjögren’s syndrome (SS), is characterized by lymphocytic infiltration and loss of function of exocrine glands such as the lacrimal gland (LG) and salivary gland. SS-associated changes in the LG are associated with the development of autoimmune-mediated dry eye disease. We have previously reported the accumulation of intercellular adhesion molecule 1 (ICAM-1) in the LG of Non-Obese Diabetic (NOD) mice, a murine model of autoimmune-mediated dry eye in SS, in both LG acinar cells and infiltrating lymphocytes. ICAM-1 initiates T-cell activation and can trigger T-cell migration through binding to lymphocyte function-associated 1 antigen (LFA). To modulate this interaction, this study introduces a new tool, a multivalent biopolymeric nanoparticle assembled from a diblock elastin-like polypeptide (ELP) using the S48I48 (SI) ELP scaffold fused with a mouse ICAM-1 targeting peptide to form IBP-SI. IBP-SI forms a multivalent, monodisperse nanoparticle with a radius of 21.9 nm. Unlike the parent SI, IBP-SI binds mouse ICAM-1 and is internalized by endocytosis into transfected HeLa cells before it accumulates in lysosomes. In vitro assays measuring lymphocyte adhesion to Tumor Necrosis Factor TNF-α-treated bEnd.3 cells, which express high levels of ICAM-1, show that adhesion is inhibited by IBP-SI but not by SI, with IC50 values of 62.7 μM and 81.2 μM, respectively, in two different assay formats. IBP-SI, but not SI, also blocked T-cell proliferation in a mixed lymphocyte reaction by 74% relative to proliferation in an untreated mixed cell reaction. These data suggest that a biopolymeric nanoparticle with affinity for ICAM-1 can disrupt ICAM-1 and LFA interactions in vitro and may have further utility as an in vivo tool or potential therapeutic.
Collapse
Affiliation(s)
- Pang-Yu Hsueh
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; (P.-Y.H.); (Y.J.); (A.V.)
| | - Yaping Ju
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; (P.-Y.H.); (Y.J.); (A.V.)
| | - Adrianna Vega
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; (P.-Y.H.); (Y.J.); (A.V.)
| | - Maria C. Edman
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA 90033, USA;
| | - J. Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; (P.-Y.H.); (Y.J.); (A.V.)
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence: (J.A.M.); (S.F.H.-A.)
| | - Sarah F. Hamm-Alvarez
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; (P.-Y.H.); (Y.J.); (A.V.)
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA 90033, USA;
- Correspondence: (J.A.M.); (S.F.H.-A.)
| |
Collapse
|
24
|
Beldman T, Malinova TS, Desclos E, Grootemaat AE, Misiak ALS, van der Velden S, van Roomen CPAA, Beckers L, van Veen HA, Krawczyk PM, Hoebe RA, Sluimer JC, Neele AE, de Winther MPJ, van der Wel NN, Lutgens E, Mulder WJM, Huveneers S, Kluza E. Nanoparticle-Aided Characterization of Arterial Endothelial Architecture during Atherosclerosis Progression and Metabolic Therapy. ACS NANO 2019; 13:13759-13774. [PMID: 31268670 PMCID: PMC6933811 DOI: 10.1021/acsnano.8b08875] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 07/03/2019] [Indexed: 05/08/2023]
Abstract
Atherosclerosis is associated with a compromised endothelial barrier, facilitating the accumulation of immune cells and macromolecules in atherosclerotic lesions. In this study, we investigate endothelial barrier integrity and the enhanced permeability and retention (EPR) effect during atherosclerosis progression and therapy in Apoe-/- mice using hyaluronan nanoparticles (HA-NPs). Utilizing ultrastructural and en face plaque imaging, we uncover a significantly decreased junction continuity in the atherosclerotic plaque-covering endothelium compared to the normal vessel wall, indicative of disrupted endothelial barrier. Intriguingly, the plaque advancement had a positive effect on junction stabilization, which correlated with a 3-fold lower accumulation of in vivo administrated HA-NPs in advanced plaques compared to early counterparts. Furthermore, by using super-resolution and correlative light and electron microscopy, we trace nanoparticles in the plaque microenvironment. We find nanoparticle-enriched endothelial junctions, containing 75% of detected HA-NPs, and a high HA-NP accumulation in the endothelium-underlying extracellular matrix, which suggest an endothelial junctional traffic of HA-NPs to the plague. Finally, we probe the EPR effect by HA-NPs in the context of metabolic therapy with a glycolysis inhibitor, 3PO, proposed as a vascular normalizing strategy. The observed trend of attenuated HA-NP uptake in aortas of 3PO-treated mice coincides with the endothelial silencing activity of 3PO, demonstrated in vitro. Interestingly, the therapy also reduced the plaque inflammatory burden, while activating macrophage metabolism. Our findings shed light on natural limitations of nanoparticle accumulation in atherosclerotic plaques and provide mechanistic insight into nanoparticle trafficking across the atherosclerotic endothelium. Furthermore, our data contribute to the rising field of endothelial barrier modulation in atherosclerosis.
Collapse
Affiliation(s)
- Thijs
J. Beldman
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Tsveta S. Malinova
- Vascular
Microenvironment and Integrity, Department of Medical Biochemistry,
Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Center, Amsterdam 1105 AZ, The
Netherlands
| | - Emilie Desclos
- Cellular
Imaging-Core Facility, Academic Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Anita E. Grootemaat
- Cellular
Imaging-Core Facility, Academic Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Aresh L. S. Misiak
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Saskia van der Velden
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Cindy P. A. A. van Roomen
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Linda Beckers
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Henk A. van Veen
- Cellular
Imaging-Core Facility, Academic Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Przemyslaw M. Krawczyk
- Department
of Medical Biology, Amsterdam University
Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Ron A. Hoebe
- Cellular
Imaging-Core Facility, Academic Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Judith C. Sluimer
- Department
of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht 6229 ER, The Netherlands
| | - Annette E. Neele
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Menno P. J. de Winther
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
- Institute
for Cardiovascular Prevention, Ludwig Maximilians
University, Munich 80336, Germany
| | - Nicole N. van der Wel
- Cellular
Imaging-Core Facility, Academic Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Esther Lutgens
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
- Institute
for Cardiovascular Prevention, Ludwig Maximilians
University, Munich 80336, Germany
| | - Willem J. M. Mulder
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
- Translational
and Molecular Imaging Institute, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Stephan Huveneers
- Vascular
Microenvironment and Integrity, Department of Medical Biochemistry,
Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Center, Amsterdam 1105 AZ, The
Netherlands
| | - Ewelina Kluza
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| |
Collapse
|
25
|
Myerson JW, McPherson O, DeFrates KG, Towslee JH, Marcos-Contreras OA, Shuvaev VV, Braender B, Composto RJ, Muzykantov VR, Eckmann DM. Cross-linker-Modulated Nanogel Flexibility Correlates with Tunable Targeting to a Sterically Impeded Endothelial Marker. ACS NANO 2019; 13:11409-11421. [PMID: 31600053 PMCID: PMC7393972 DOI: 10.1021/acsnano.9b04789] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Deformability of injectable nanocarriers impacts rheological behavior, drug loading, and affinity target adhesion. Here, we present atomic force microscopy (AFM) and spectroscopy measurements of nanocarrier Young's moduli, tune the moduli of deformable nanocarriers with cross-linkers, and demonstrate vascular targeting behavior that correlates with Young's modulus. Homobifunctional cross-linkers were introduced into lysozyme-dextran nanogels (NGs). Single particle-scale AFM measurements determined NG moduli varying from ∼50-150 kPa for unmodified NGs or NGs with a short hydrophilic cross-linker (2,2'-(ethylenedioxy)bis(ethylamine), EOD) to ∼350 kPa for NGs modified with a longer hydrophilic cross-linker (4,9-dioxa-1,12-dodecanediamine, DODD) to ∼10 MPa for NGs modified with a longer hydrophobic cross-linker (1,12-diaminododecane, DAD). Cross-linked NGs were conjugated to antibodies for plasmalemma vesicle associated protein (PLVAP), a caveolar endothelial marker that cannot be accessed by rigid particles larger than ∼100 nm. In previous work, 150 nm NGs effectively targeted PLVAP, where rigid particles of similar diameter did not. EOD-modified NGs targeted PLVAP less effectively than unmodified NGs, but more effectively than DODD or DAD modified NGs, which both yielded low levels of targeting, resembling results previously obtained with polystyrene particles. Cross-linked NGs were also conjugated to antibodies against intracellular adhesion molecule-1 (ICAM-1), an endothelial marker accessible to large rigid particles. Cross-linked NGs and unmodified NGs targeted uniformly to ICAM-1. We thus demonstrate cross-linker modification of NGs, AFM determination of NG mechanical properties varying with cross-linker, and tuning of specific sterically constrained vascular targeting behavior in correlation with cross-linker-modified NG mechanical properties.
Collapse
Affiliation(s)
- Jacob Wheatley Myerson
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Olivia McPherson
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Kelsey G. DeFrates
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jenna H. Towslee
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A. Marcos-Contreras
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir V. Shuvaev
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Bruce Braender
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Russell J. Composto
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R. Muzykantov
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Corresponding Author:
| | - David M. Eckmann
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
26
|
Mahmoud AM, de Jongh PAJM, Briere S, Chen M, Nowell CJ, Johnston APR, Davis TP, Haddleton DM, Kempe K. Carboxylated Cy5-Labeled Comb Polymers Passively Diffuse the Cell Membrane and Target Mitochondria. ACS APPLIED MATERIALS & INTERFACES 2019; 11:31302-31310. [PMID: 31369228 DOI: 10.1021/acsami.9b09395] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A detailed understanding of the cellular uptake and trafficking of nanomaterials is essential for the design of "smart" intracellular drug delivery vehicles. Typically, cellular interactions can be tailored by endowing materials with specific properties, for example, through the introduction of charges or targeting groups. In this study, water-soluble carboxylated N-acylated poly(amino ester)-based comb polymers of different degree of polymerization and side-chain modification were synthesized via a combination of spontaneous zwitterionic copolymerization and redox-initiated reversible addition-fragmentation chain-transfer polymerization and fully characterized by 1H NMR spectroscopy and size exclusion chromatography. The comb polymers showed no cell toxicity against NIH/3T3 and N27 cell lines nor hemolysis. Detailed cellular association and uptake studies by flow cytometry and confocal laser scanning microscopy (CLSM) revealed that the carboxylated polymers were capable of passively diffusing cell membranes and targeting mitochondria. The interplay of pendant carboxylic acids of the comb polymers and the Cy5-label was identified as major driving force for this behavior, which was demonstrated to be applicable in NIH/3T3 and N27 cell lines. Blocking of the carboxylic acids through modification with 2-methoxyethylamine and poly(2-ethyl-2-oxazoline) or replacement of the dye label with a different dye (e.g., fluorescein) resulted in an alteration of the cellular uptake mechanism toward endocytosis as demonstrated by CLSM. In contrast, partial modification of the carboxylic acid groups allowed to retain the cellular interaction, hence, rendering these comb polymers a highly functional mitochondria targeted carrier platform for future drug delivery applications and imaging purposes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - David M Haddleton
- Department of Chemistry , University of Warwick , Coventry CV4 7AL , United Kingdom
| | | |
Collapse
|
27
|
Ju Y, Guo H, Yarber F, Edman MC, Peddi S, Janga SR, MacKay JA, Hamm-Alvarez SF. Molecular Targeting of Immunosuppressants Using a Bifunctional Elastin-Like Polypeptide. Bioconjug Chem 2019; 30:2358-2372. [PMID: 31408605 DOI: 10.1021/acs.bioconjchem.9b00462] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Elastin-Like Polypeptides (ELP) are environmentally responsive protein polymers which are easy to engineer and biocompatible, making them ideal candidates as drug carriers. Our team has recently utilized ELPs fused to FKBP12 to carry Rapamycin (Rapa), a potent immunosuppressant. Through high affinity binding to Rapa, FKBP carriers can yield beneficial therapeutic effects and reduce the off-site toxicity of Rapa. Since ICAM-1 is significantly elevated at sites of inflammation in diverse diseases, we hypothesized that a molecularly targeted ELP carrier capable of binding ICAM-1 might have advantageous properties. Here we report on the design, characterization, pharmacokinetics, and biodistribution of a new ICAM-1-targeted ELP Rapa carrier (IBPAF) and its preliminary characterization in a murine model exhibiting elevated ICAM-1. Lacrimal glands (LG) of male NOD mice, a disease model recapitulating the autoimmune dacryoadenitis seen in Sjögren's Syndrome patients, were analyzed to confirm that ICAM-1 was significantly elevated in the LG relative to control male BALB/c mice (3.5-fold, p < 0.05, n = 6). In vitro studies showed that IBPAF had significantly higher binding to TNF-α-stimulated bEnd.3 cells which overexpress surface ICAM-1, relative to nontargeted control ELP (AF)(4.0-fold, p < 0.05). A pharmacokinetics study in male NOD mice showed no significant differences between AF and IBPAF for plasma half-life, clearance, and volume of distribution. However, both constructs maintained a higher level of Rapa in systemic circulation compared to free Rapa. Interestingly, in the male NOD mouse, the accumulation of IBPAF was significantly higher in homogenized LG extracts compared to AF at 2 h (8.6 ± 6.6% versus 1.3 ± 1.3%, respectively, n = 5, p < 0.05). This accumulation was transient with no differences detected at 8 or 24 h. This study describes the first ICAM-1 targeted protein-polymer carrier for Rapa that specifically binds to ICAM-1 in vitro and accumulates in ICAM-1 overexpressing tissue in vivo, which may be useful for molecular targeting in diverse inflammatory diseases where ICAM-1 is elevated.
Collapse
Affiliation(s)
- Yaping Ju
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy , University of Southern California , Los Angeles , California 90089 , United States.,Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine , University of Southern California , Los Angeles , California 90089 , United States
| | - Hao Guo
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy , University of Southern California , Los Angeles , California 90089 , United States
| | - Frances Yarber
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine , University of Southern California , Los Angeles , California 90089 , United States
| | - Maria C Edman
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine , University of Southern California , Los Angeles , California 90089 , United States
| | - Santosh Peddi
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy , University of Southern California , Los Angeles , California 90089 , United States
| | - Srikanth Reddy Janga
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine , University of Southern California , Los Angeles , California 90089 , United States
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy , University of Southern California , Los Angeles , California 90089 , United States.,Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine , University of Southern California , Los Angeles , California 90089 , United States.,Department of Biomedical Engineering, Viterbi School of Engineering , University of Southern California , Los Angeles , California 90089 , United States
| | - Sarah F Hamm-Alvarez
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy , University of Southern California , Los Angeles , California 90089 , United States.,Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine , University of Southern California , Los Angeles , California 90089 , United States
| |
Collapse
|
28
|
Manthe RL, Rappaport JA, Long Y, Solomon M, Veluvolu V, Hildreth M, Gugutkov D, Marugan J, Zheng W, Muro S. δ-Tocopherol Effect on Endocytosis and Its Combination with Enzyme Replacement Therapy for Lysosomal Disorders: A New Type of Drug Interaction? J Pharmacol Exp Ther 2019; 370:823-833. [PMID: 31101681 DOI: 10.1124/jpet.119.257345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/15/2019] [Indexed: 12/27/2022] Open
Abstract
Induction of lysosomal exocytosis alleviates lysosomal storage of undigested metabolites in cell models of lysosomal disorders (LDs). However, whether this strategy affects other vesicular compartments, e.g., those involved in endocytosis, is unknown. This is important both to predict side effects and to use this strategy in combination with therapies that require endocytosis for intracellular delivery, such as lysosomal enzyme replacement therapy (ERT). We investigated this using δ-tocopherol as a model previously shown to induce lysosomal exocytosis and cell models of type A Niemann-Pick disease, a LD characterized by acid sphingomyelinase (ASM) deficiency and sphingomyelin storage. δ-Tocopherol and derivative CF3-T reduced net accumulation of fluid phase, ligands, and polymer particles via phagocytic, caveolae-, clathrin-, and cell adhesion molecule (CAM)-mediated pathways, yet the latter route was less affected due to receptor overexpression. In agreement, δ-tocopherol lowered uptake of recombinant ASM by deficient cells (known to occur via the clathrin pathway) and via targeting intercellular adhesion molecule-1 (associated to the CAM pathway). However, the net enzyme activity delivered and lysosomal storage attenuation were greater via the latter route. Data suggest stimulation of exocytosis by tocopherols is not specific of lysosomes and affects endocytic cargo. However, this effect was transient and became unnoticeable several hours after tocopherol removal. Therefore, induction of exocytosis in combination with therapies requiring endocytic uptake, such as ERT, may represent a new type of drug interaction, yet this strategy could be valuable if properly timed for minimal interference.
Collapse
Affiliation(s)
- Rachel L Manthe
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Jeffrey A Rappaport
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Yan Long
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Melani Solomon
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Vinay Veluvolu
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Michael Hildreth
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Dencho Gugutkov
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Juan Marugan
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Wei Zheng
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Silvia Muro
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| |
Collapse
|
29
|
Lai J, Deng G, Sun Z, Peng X, Li J, Gong P, Zhang P, Cai L. Scaffolds biomimicking macrophages for a glioblastoma NIR-Ib imaging guided photothermal therapeutic strategy by crossing Blood-Brain Barrier. Biomaterials 2019; 211:48-56. [PMID: 31085358 DOI: 10.1016/j.biomaterials.2019.04.026] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/15/2019] [Accepted: 04/23/2019] [Indexed: 12/22/2022]
Abstract
Glioblastoma (GBM) is one of the most malignant cancers, and Blood-Brain Barrier (BBB) is the main obstacle to diagnose and treat GBM, hence scientists are making great efforts to develop new drugs which can pass BBB and integrate diagnosis and therapeutics together. Here, we designed plasma membrane of macrophage camouflaged DSPE-PEG loaded near-infrared Ib (NIR-Ib) fluorescence dye IR-792 nanoparticles (MDINPs). MDINPs were able to penetrate BBB and selectively accumulate at tumor site, and then could be used as NIR-Ib fluorescence probes for targeted tumor imaging. At the same time, MDINPs could kill tumor cells by photothermal effect. Our results showed that MDINPs-mediated NIR-Ib fluorescence imaging could clearly observe orthotopic GBM, and the NIR-Ib imaging-guided photothermal therapy significantly suppressed the growth of GBM and prolonged the life of mice. This work not only provided a method to mimic the biological function of macrophage, but also provided an integrative strategy for diagnosis and treatment in GBM.
Collapse
Affiliation(s)
- Jiazheng Lai
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Guanjun Deng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zhihong Sun
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xinghua Peng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jing Li
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ping Gong
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Dongguan, 523808, China.
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Chemical and Biological Engineering, Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction and Institute for Advanced Study, The Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Kowloon, Hong Kong, China.
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
30
|
Cardon F, Pallisse R, Bardor M, Caron A, Vanier J, Ele Ekouna JP, Lerouge P, Boitel‐Conti M, Guillet M. Brassica rapa hairy root based expression system leads to the production of highly homogenous and reproducible profiles of recombinant human alpha-L-iduronidase. PLANT BIOTECHNOLOGY JOURNAL 2019; 17:505-516. [PMID: 30058762 PMCID: PMC6335068 DOI: 10.1111/pbi.12994] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/02/2018] [Accepted: 07/22/2018] [Indexed: 05/28/2023]
Abstract
The Brassica rapa hairy root based expression platform, a turnip hairy root based expression system, is able to produce human complex glycoproteins such as the alpha-L-iduronidase (IDUA) with an activity similar to the one produced by Chinese Hamster Ovary (CHO) cells. In this article, a particular attention has been paid to the N- and O-glycosylation that characterize the alpha-L-iduronidase produced using this hairy root based system. This analysis showed that the recombinant protein is characterized by highly homogeneous post translational profiles enabling a strong batch to batch reproducibility. Indeed, on each of the 6 N-glycosylation sites of the IDUA, a single N-glycan composed of a core Man3 GlcNAc2 carrying one beta(1,2)-xylose and one alpha(1,3)-fucose epitope (M3XFGN2) was identified, highlighting the high homogeneity of the production system. Hydroxylation of proline residues and arabinosylation were identified during O-glycosylation analysis, still with a remarkable reproducibility. This platform is thus positioned as an effective and consistent expression system for the production of human complex therapeutic proteins.
Collapse
Affiliation(s)
| | | | - Muriel Bardor
- Laboratoire Glyco‐MEV EA4358UNIROUENNormandie UniversitéRouenFrance
- Institut Universitaire de France (I.U.F.)Paris Cedex 05France
| | | | - Jessica Vanier
- Laboratoire Glyco‐MEV EA4358UNIROUENNormandie UniversitéRouenFrance
| | - Jean Pierre Ele Ekouna
- Biologie des Plantes et Innovation (BIOPI)Université de Picardie Jules VerneAmiensFrance
| | - Patrice Lerouge
- Laboratoire Glyco‐MEV EA4358UNIROUENNormandie UniversitéRouenFrance
| | - Michèle Boitel‐Conti
- Biologie des Plantes et Innovation (BIOPI)Université de Picardie Jules VerneAmiensFrance
| | | |
Collapse
|
31
|
Muro S. Alterations in Cellular Processes Involving Vesicular Trafficking and Implications in Drug Delivery. Biomimetics (Basel) 2018; 3:biomimetics3030019. [PMID: 31105241 PMCID: PMC6352689 DOI: 10.3390/biomimetics3030019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/31/2022] Open
Abstract
Endocytosis and vesicular trafficking are cellular processes that regulate numerous functions required to sustain life. From a translational perspective, they offer avenues to improve the access of therapeutic drugs across cellular barriers that separate body compartments and into diseased cells. However, the fact that many factors have the potential to alter these routes, impacting our ability to effectively exploit them, is often overlooked. Altered vesicular transport may arise from the molecular defects underlying the pathological syndrome which we aim to treat, the activity of the drugs being used, or side effects derived from the drug carriers employed. In addition, most cellular models currently available do not properly reflect key physiological parameters of the biological environment in the body, hindering translational progress. This article offers a critical overview of these topics, discussing current achievements, limitations and future perspectives on the use of vesicular transport for drug delivery applications.
Collapse
Affiliation(s)
- Silvia Muro
- Institute for Bioscience and Biotechnology Research and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain.
- Institute for Bioengineering of Catalonia (IBEC) of the Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain.
| |
Collapse
|
32
|
The Multifaceted Uses and Therapeutic Advantages of Nanoparticles for Atherosclerosis Research. MATERIALS 2018; 11:ma11050754. [PMID: 29738480 PMCID: PMC5978131 DOI: 10.3390/ma11050754] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 12/27/2022]
Abstract
Nanoparticles are uniquely suited for the study and development of potential therapies against atherosclerosis by virtue of their size, fine-tunable properties, and ability to incorporate therapies and/or imaging modalities. Furthermore, nanoparticles can be specifically targeted to the atherosclerotic plaque, evading off-target effects and/or associated cytotoxicity. There has been a wealth of knowledge available concerning the use of nanotechnologies in cardiovascular disease and atherosclerosis, in particular in animal models, but with a major focus on imaging agents. In fact, roughly 60% of articles from an initial search for this review included examples of imaging applications of nanoparticles. Thus, this review focuses on experimental therapy interventions applied to and observed in animal models. Particular emphasis is placed on how nanoparticle materials and properties allow researchers to learn a great deal about atherosclerosis. The objective of this review was to provide an update for nanoparticle use in imaging and drug delivery studies and to illustrate how nanoparticles can be used for sensing and modelling, for studying fundamental biological mechanisms, and for the delivery of biotherapeutics such as proteins, peptides, nucleic acids, and even cells all with the goal of attenuating atherosclerosis. Furthermore, the various atherosclerosis processes targeted mainly for imaging studies have been summarized in the hopes of inspiring new and exciting targeted therapeutic and/or imaging strategies.
Collapse
|
33
|
Sharma A, Vaghasiya K, Ray E, Verma RK. Lysosomal targeting strategies for design and delivery of bioactive for therapeutic interventions. J Drug Target 2017; 26:208-221. [DOI: 10.1080/1061186x.2017.1374390] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Ankur Sharma
- Institute of Nano Science and Technology (INST), Phase 10, Mohali, India
| | - Kalpesh Vaghasiya
- Institute of Nano Science and Technology (INST), Phase 10, Mohali, India
| | - Eupa Ray
- Institute of Nano Science and Technology (INST), Phase 10, Mohali, India
| | - Rahul Kumar Verma
- Institute of Nano Science and Technology (INST), Phase 10, Mohali, India
| |
Collapse
|
34
|
Solomon M, Muro S. Lysosomal enzyme replacement therapies: Historical development, clinical outcomes, and future perspectives. Adv Drug Deliv Rev 2017; 118:109-134. [PMID: 28502768 PMCID: PMC5828774 DOI: 10.1016/j.addr.2017.05.004] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 01/06/2023]
Abstract
Lysosomes and lysosomal enzymes play a central role in numerous cellular processes, including cellular nutrition, recycling, signaling, defense, and cell death. Genetic deficiencies of lysosomal components, most commonly enzymes, are known as "lysosomal storage disorders" or "lysosomal diseases" (LDs) and lead to lysosomal dysfunction. LDs broadly affect peripheral organs and the central nervous system (CNS), debilitating patients and frequently causing fatality. Among other approaches, enzyme replacement therapy (ERT) has advanced to the clinic and represents a beneficial strategy for 8 out of the 50-60 known LDs. However, despite its value, current ERT suffers from several shortcomings, including various side effects, development of "resistance", and suboptimal delivery throughout the body, particularly to the CNS, lowering the therapeutic outcome and precluding the use of this strategy for a majority of LDs. This review offers an overview of the biomedical causes of LDs, their socio-medical relevance, treatment modalities and caveats, experimental alternatives, and future treatment perspectives.
Collapse
Affiliation(s)
- Melani Solomon
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University Maryland, College Park, MD 20742, USA.
| |
Collapse
|
35
|
Kim SW, Lee YK, Kim SH, Park JY, Lee DU, Choi J, Hong JH, Kim S, Khang D. Covalent, Non-Covalent, Encapsulated Nanodrug Regulate the Fate of Intra- and Extracellular Trafficking: Impact on Cancer and Normal Cells. Sci Rep 2017; 7:6454. [PMID: 28743942 PMCID: PMC5526881 DOI: 10.1038/s41598-017-06796-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/28/2017] [Indexed: 11/20/2022] Open
Abstract
Drugs need to be designed to access the designated intracellular organelle compartments in order to maximize anticancer efficacy. This study identified that covalently conjugated, non-covalent polyethylene glycol coated and encapsulated nanodrugs selectively influence drug uptake, the intracellular and extracellular trafficking of cancer cells. The types of nano conjugation modulated intracellular dynamics associated with differential impact on anti-cancer efficacy, but also induced differential cytotoxicity on cancer versus normal cells. In conclusion, this study demonstrated the importance of selecting the appropriate type of nano-conjugation for delivering organelle specific, active chemotherapeutic agents through controlled intracellular trafficking.
Collapse
Affiliation(s)
- Sang-Woo Kim
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Yeon Kyung Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Sang-Hyun Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, 41566, South Korea
| | - Jun-Young Park
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Dong Un Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Jungil Choi
- Gyeongnam Department of Environment Toxicology and Chemistry, Korea Institutes of Toxicology, Jinju, 52834, South Korea
| | - Jeong Hee Hong
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea.,Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, South Korea
| | - Sanghyo Kim
- Department of Bionanotechnology, Gachon University, Seongnam, 13120, South Korea.
| | - Dongwoo Khang
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea. .,Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, South Korea.
| |
Collapse
|
36
|
Garnacho C, Muro S. ICAM-1 targeting, intracellular trafficking, and functional activity of polymer nanocarriers coated with a fibrinogen-derived peptide for lysosomal enzyme replacement. J Drug Target 2017; 25:786-795. [PMID: 28665212 DOI: 10.1080/1061186x.2017.1349771] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Enzyme replacement is a viable treatment for diseases caused by genetic deficiency of lysosomal enzymes. However, suboptimal access of enzymes to target sites limits this strategy. Polymer nanocarriers (NCs) coated with antibody against intercellular adhesion molecule 1 (ICAM-1), a protein overexpressed on most cells under disease states, enhanced biodistribution and lysosomal delivery of these therapeutics. Whether this can be achieved using more biocompatible ICAM-1-targeting moieties is unknown, since intracellular uptake via this route is sensitive to the receptor epitope being targeted. We examined this using polymer NCs coated with an ICAM-1-targeting peptide derived from the fibrinogen sequence. Scrambled-sequence peptide and anti-ICAM were used as controls. NCs carried acid sphingomyelinase (ASM), used for treatment of type B Niemann-Pick disease, and fluorescence microscopy was employed to examine cellular performance. Peptide-coated/enzyme NCs efficiently targeted ICAM-1 (22-fold over non-specific counterparts; Bmax ∼180 NCs/cell; t1/2 ∼28 min), recognised human and mouse cells (1.2- to 0.7-fold binding vs. antibody/enzyme NCs), were efficiently endocytosed (71% at 1 h chase), and trafficked to lysosomes (30--45% of internalised NCs; 2 h chase). This restored lysosomal levels of sphingomyelin and cholesterol within 5 h chase (∼95% reduction over disease levels), similar to antibody-enzyme NCs. This fibrinogen-derived ICAM-1-targeting peptide holds potential for lysosomal enzyme replacement therapy.
Collapse
Affiliation(s)
- Carmen Garnacho
- a Department of Normal and Pathological Histology and Cytology , University of Seville School of Medicine , Seville , Spain
| | - Silvia Muro
- b Institute for Bioscience & Biotechnology Research, University of Maryland , College Park , MD , USA.,c Fischell Department of Bioengineering , University of Maryland , College Park , MD , USA
| |
Collapse
|
37
|
Okamoto Y, Shikano S. Differential phosphorylation signals control endocytosis of GPR15. Mol Biol Cell 2017; 28:2267-2281. [PMID: 28615320 PMCID: PMC5555655 DOI: 10.1091/mbc.e16-09-0627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 12/30/2022] Open
Abstract
GPR15 undergoes a ligand-independent endocytosis, which requires phosphorylation of a distal C-terminal Ser-357 mediated by multiple basophilic kinases. The functional role of Ser-357 in endocytosis is distinct from that of a conserved Ser/Thr cluster, which is more responsible for the use of GRKs and β-arrestin. GPR15 is an orphan G protein–coupled receptor (GPCR) that serves for an HIV coreceptor and was also recently found as a novel homing receptor for T-cells implicated in colitis. We show that GPR15 undergoes a constitutive endocytosis in the absence of ligand. The endocytosis was clathrin dependent and partially dependent on β-arrestin in HEK293 cells, and nearly half of the internalized GPR15 receptors were recycled to the plasma membrane. An Ala mutation of the distal C-terminal Arg-354 or Ser-357, which forms a consensus phosphorylation site for basophilic kinases, markedly reduced the endocytosis, whereas phosphomimetic mutation of Ser-357 to Asp did not. Ser-357 was phosphorylated in vitro by multiple kinases, including PKA and PKC, and pharmacological activation of these kinases enhanced both phosphorylation of Ser-357 and endocytosis of GPR15. These results suggested that Ser-357 phosphorylation critically controls the ligand-independent endocytosis of GPR15. The functional role of Ser-357 in endocytosis was distinct from that of a conserved Ser/Thr cluster in the more proximal C-terminus, which was responsible for the β-arrestin– and GPCR kinase–dependent endocytosis of GPR15. Thus phosphorylation signals may differentially control cell surface density of GPR15 through endocytosis.
Collapse
Affiliation(s)
- Yukari Okamoto
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607-7170
| | - Sojin Shikano
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607-7170
| |
Collapse
|
38
|
Garnacho C, Dhami R, Solomon M, Schuchman EH, Muro S. Enhanced Delivery and Effects of Acid Sphingomyelinase by ICAM-1-Targeted Nanocarriers in Type B Niemann-Pick Disease Mice. Mol Ther 2017; 25:1686-1696. [PMID: 28606376 DOI: 10.1016/j.ymthe.2017.05.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 05/19/2017] [Accepted: 05/21/2017] [Indexed: 01/01/2023] Open
Abstract
Acid sphingomyelinase deficiency in type B Niemann-Pick disease leads to lysosomal sphingomyelin storage, principally affecting lungs, liver, and spleen. Infused recombinant enzyme is beneficial, yet its delivery to the lungs is limited and requires higher dosing than liver and spleen, leading to potentially adverse reactions. Previous studies showed increased enzyme pulmonary uptake by nanocarriers targeted to ICAM-1, a protein overexpressed during inflammation. Here, using polystyrene and poly(lactic-co-glycolic acid) nanocarriers, we optimized lung delivery by varying enzyme dose and nanocarrier concentration, verified endocytosis and lysosomal trafficking in vivo, and evaluated delivered activity and effects. Raising the enzyme load of nanocarriers progressively increased absolute enzyme delivery to all lung, liver, and spleen, over the naked enzyme. Varying nanocarrier concentration inversely impacted lung versus liver and spleen uptake. Mouse intravital and postmortem examination verified endocytosis, transcytosis, and lysosomal trafficking using nanocarriers. Compared to naked enzyme, nanocarriers increased enzyme activity in organs and reduced lung sphingomyelin storage and macrophage infiltration. Although old mice with advanced disease showed reactivity (pulmonary leukocyte infiltration) to injections, including buffer without carriers, antibody, or enzyme, younger mice with mild disease did not. We conclude that anti-ICAM nanocarriers may result in effective lung enzyme therapy using low enzyme doses.
Collapse
Affiliation(s)
- Carmen Garnacho
- Department of Normal and Pathological Histology and Cytology, University of Seville School of Medicine, 41009 Seville, Spain
| | - Rajwinder Dhami
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Melani Solomon
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA
| | - Edward H Schuchman
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
39
|
Schuster T, Mühlstein A, Yaghootfam C, Maksimenko O, Shipulo E, Gelperina S, Kreuter J, Gieselmann V, Matzner U. Potential of surfactant-coated nanoparticles to improve brain delivery of arylsulfatase A. J Control Release 2017; 253:1-10. [DOI: 10.1016/j.jconrel.2017.02.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/10/2017] [Accepted: 02/15/2017] [Indexed: 02/08/2023]
|
40
|
Abstract
Nanoparticles, structures of less than 200 nm capable of delivering pharmacotherapeutics to sites of disease, have shown great promise for the treatment of many disease states. While no nanoparticle therapies for deep vein thrombosis are currently approved by the Food and Drug Administration, many of the unique features of these therapies have the potential to treat both deep vein thrombosis and its most significant sequela, postthrombotic syndrome, while limiting the hemorrhagic complications of current antithrombotic therapies. Nanoparticles are complex structures with several important variables that must be considered to engineer effective therapies. This article will review the structure and engineering of nanoparticles, as well as promising molecular targets for future investigation.
Collapse
Affiliation(s)
- Benjamin Jacobs
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Chandu Vemuri
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
41
|
Fliervoet LAL, Mastrobattista E. Drug delivery with living cells. Adv Drug Deliv Rev 2016; 106:63-72. [PMID: 27129442 DOI: 10.1016/j.addr.2016.04.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 12/25/2022]
Abstract
The field of drug delivery has grown tremendously in the past few decades by developing a wide range of advanced drug delivery systems. An interesting category is cell-based drug delivery, which includes encapsulation of drugs inside cells or attached to the surface and subsequent transportation through the body. Another approach involves genetic engineering of cells to secrete therapeutic molecules in a controlled way. The next-generation systems integrate expertise from synthetic biology to generate therapeutic gene networks for highly advanced sensory and output devices. These developments are very exciting for the drug delivery field and could radically change the way we administer biological medicines to chronically ill patients. This review is covering the use of living cells, either as transport system or production-unit, to deliver therapeutic molecules and bioactive proteins inside the body. It describes a wide range of approaches in cell-based drug delivery and highlights exceptional examples.
Collapse
Affiliation(s)
- Lies A L Fliervoet
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands.
| |
Collapse
|
42
|
Giannotti MI, Abasolo I, Oliva M, Andrade F, García-Aranda N, Melgarejo M, Pulido D, Corchero JL, Fernández Y, Villaverde A, Royo M, García-Parajo MF, Sanz F, Schwartz S. Highly Versatile Polyelectrolyte Complexes for Improving the Enzyme Replacement Therapy of Lysosomal Storage Disorders. ACS APPLIED MATERIALS & INTERFACES 2016; 8:25741-25752. [PMID: 27610822 DOI: 10.1021/acsami.6b08356] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Lysosomal storage disorders are currently treated by enzyme replacement therapy (ERT) through the direct administration of the unprotected recombinant protein to the patients. Herein we present an ionically cross-linked polyelectrolyte complex (PEC) composed of trimethyl chitosan (TMC) and α-galactosidase A (GLA), the defective enzyme in Fabry disease, with the capability of directly targeting endothelial cells by incorporating peptide ligands containing the RGD sequence. We assessed the physicochemical properties, cytotoxicity, and hemocompatibility of RGD-targeted and untargeted PECs, the uptake by endothelial cells and the intracellular activity of PECs in cell culture models of Fabry disease. Moreover, we also explored the effect of different freeze-drying procedures in the overall activity of the PECs. Our results indicate that the use of integrin-binding RGD moiety within the PEC increases their uptake and the efficacy of the GLA enzyme, while the freeze-drying allows the activity of the therapeutic protein to remain intact. Overall, these results highlight the potential of TMC-based PECs as a highly versatile and feasible drug delivery system for improving the ERT of lysosomal storage disorders.
Collapse
Affiliation(s)
- Marina I Giannotti
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Physical Chemistry Department, Universitat de Barcelona , 08028 Barcelona, Spain
| | - Ibane Abasolo
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| | - Mireia Oliva
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Pharmacy and Pharmaceutical Technology Department, Universitat de Barcelona , 08028 Barcelona, Spain
| | - Fernanda Andrade
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Pharmacy and Pharmaceutical Technology Department, Universitat de Barcelona , 08028 Barcelona, Spain
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto , 4050-313 Porto, Portugal
| | - Natalia García-Aranda
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| | - Marta Melgarejo
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Combinatorial Chemistry Unit, Barcelona Science Park , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Daniel Pulido
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Combinatorial Chemistry Unit, Barcelona Science Park , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - José L Corchero
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona , Bellaterra, 08193 Cerdanyola del Vallès, Spain
| | - Yolanda Fernández
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| | - Antonio Villaverde
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona , Bellaterra, 08193 Cerdanyola del Vallès, Spain
| | - Miriam Royo
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Combinatorial Chemistry Unit, Barcelona Science Park , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - María F García-Parajo
- Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology , ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Fausto Sanz
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Physical Chemistry Department, Universitat de Barcelona , 08028 Barcelona, Spain
| | - Simó Schwartz
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| |
Collapse
|
43
|
Long Y, Xu M, Li R, Dai S, Beers J, Chen G, Soheilian F, Baxa U, Wang M, Marugan JJ, Muro S, Li Z, Brady R, Zheng W. Induced Pluripotent Stem Cells for Disease Modeling and Evaluation of Therapeutics for Niemann-Pick Disease Type A. Stem Cells Transl Med 2016; 5:1644-1655. [PMID: 27484861 PMCID: PMC5189647 DOI: 10.5966/sctm.2015-0373] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/13/2016] [Indexed: 11/16/2022] Open
Abstract
Niemann-Pick disease type A (NPA)-induced pluripotent stem cells from patient dermal fibroblasts were differentiated into neural stem cells. By using the differentiated NPA neuronal cells as a cell-based disease model system, α-tocopherol, δ-tocopherol, and hydroxypropyl-β-cyclodextrin significantly reduced sphingomyelin accumulation in these NPA neuronal cells. This cell-based NPA model can be used for further study of disease pathophysiology and for high-throughput screening of compound libraries to identify lead compounds for drug development. Niemann-Pick disease type A (NPA) is a lysosomal storage disease caused by mutations in the SMPD1 gene that encodes acid sphingomyelinase (ASM). Deficiency in ASM function results in lysosomal accumulation of sphingomyelin and neurodegeneration. Currently, there is no effective treatment for NPA. To accelerate drug discovery for treatment of NPA, we generated induced pluripotent stem cells from two patient dermal fibroblast lines and differentiated them into neural stem cells. The NPA neural stem cells exhibit a disease phenotype of lysosomal sphingomyelin accumulation and enlarged lysosomes. By using this disease model, we also evaluated three compounds that reportedly reduced lysosomal lipid accumulation in Niemann-Pick disease type C as well as enzyme replacement therapy with ASM. We found that α-tocopherol, δ-tocopherol, hydroxypropyl-β-cyclodextrin, and ASM reduced sphingomyelin accumulation and enlarged lysosomes in NPA neural stem cells. Therefore, the NPA neural stem cells possess the characteristic NPA disease phenotype that can be ameliorated by tocopherols, cyclodextrin, and ASM. Our results demonstrate the efficacies of cyclodextrin and tocopherols in the NPA cell-based model. Our data also indicate that the NPA neural stem cells can be used as a new cell-based disease model for further study of disease pathophysiology and for high-throughput screening to identify new lead compounds for drug development. Significance Currently, there is no effective treatment for Niemann-Pick disease type A (NPA). To accelerate drug discovery for treatment of NPA, NPA-induced pluripotent stem cells were generated from patient dermal fibroblasts and differentiated into neural stem cells. By using the differentiated NPA neuronal cells as a cell-based disease model system, α-tocopherol, δ-tocopherol, and hydroxypropyl-β-cyclodextrin significantly reduced sphingomyelin accumulation in these NPA neuronal cells. Therefore, this cell-based NPA model can be used for further study of disease pathophysiology and for high-throughput screening of compound libraries to identify lead compounds for drug development.
Collapse
Affiliation(s)
- Yan Long
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Rong Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Sheng Dai
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jeanette Beers
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Guokai Chen
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
- Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| | - Ferri Soheilian
- Electron Microscopy Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Ulrich Baxa
- Electron Microscopy Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Mengqiao Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Juan J Marugan
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Maryland, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Zhiyuan Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Roscoe Brady
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
44
|
Ma X, Gong N, Zhong L, Sun J, Liang XJ. Future of nanotherapeutics: Targeting the cellular sub-organelles. Biomaterials 2016; 97:10-21. [DOI: 10.1016/j.biomaterials.2016.04.026] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/02/2016] [Accepted: 04/20/2016] [Indexed: 11/25/2022]
|
45
|
RNAi delivery by exosome-mimetic nanovesicles - Implications for targeting c-Myc in cancer. Biomaterials 2016; 102:231-8. [PMID: 27344366 DOI: 10.1016/j.biomaterials.2016.06.024] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/09/2016] [Accepted: 06/11/2016] [Indexed: 12/13/2022]
Abstract
To develop RNA-based therapeutics, it is crucial to create delivery vectors that transport the RNA molecule into the cell cytoplasm. Naturally released exosomes vesicles (also called "Extracellular Vesicles") have been proposed as possible RNAi carriers, but their yield is relatively small in any cell culture system. We have previously generated exosome-mimetic nanovesicles (NV) by serial extrusions of cells through nano-sized filters, which results in 100-times higher yield of extracellular vesicles. We here test 1) whether NV can be loaded with siRNA exogenously and endogenously, 2) whether the siRNA-loaded NV are taken up by recipient cells, and 3) whether the siRNA can induce functional knock-down responses in recipient cells. A siRNA against GFP was first loaded into NV by electroporation, or a c-Myc shRNA was expressed inside of the cells. The NV were efficiently loaded with siRNA with both techniques, were taken up by recipient cells, which resulted in attenuation of target gene expression. In conclusion, our study suggests that exosome-mimetic nanovesicles can be a platform for RNAi delivery to cell cytoplasm.
Collapse
|
46
|
Boal F, Puhar A, Xuereb JM, Kunduzova O, Sansonetti PJ, Payrastre B, Tronchère H. PI5P Triggers ICAM-1 Degradation in Shigella Infected Cells, Thus Dampening Immune Cell Recruitment. Cell Rep 2016; 14:750-759. [PMID: 26776508 DOI: 10.1016/j.celrep.2015.12.079] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/19/2015] [Accepted: 12/16/2015] [Indexed: 12/13/2022] Open
Abstract
Shigella flexneri, the pathogen responsible for bacillary dysentery, has evolved multiple strategies to control the inflammatory response. Here, we show that Shigella subverts the subcellular trafficking of the intercellular adhesion molecule-1 (ICAM-1), a key molecule in immune cell recruitment, in a mechanism dependent on the injected bacterial enzyme IpgD and its product, the lipid mediator PI5P. Overexpression of IpgD, but not a phosphatase dead mutant, induced the internalization and the degradation of ICAM-1 in intestinal epithelial cells. Remarkably, addition of permeant PI5P reproduced IpgD effects and led to the inhibition of neutrophil recruitment. Finally, these results were confirmed in an in vivo model of Shigella infection where IpgD-dependent ICAM-1 internalization reduced neutrophil adhesion. In conclusion, we describe here an immune evasion mechanism used by the pathogen Shigella to divert the host cell trafficking machinery in order to reduce immune cell recruitment.
Collapse
Affiliation(s)
- Frédéric Boal
- INSERM U1048, I2MC and Université Paul Sabatier, 31432 Toulouse, France
| | - Andrea Puhar
- INSERM U1202, Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 75724 Paris Cedex 15, France; The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR) and Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden
| | - Jean-Marie Xuereb
- INSERM U1048, I2MC and Université Paul Sabatier, 31432 Toulouse, France
| | - Oksana Kunduzova
- INSERM U1048, I2MC and Université Paul Sabatier, 31432 Toulouse, France
| | - Philippe J Sansonetti
- INSERM U1202, Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 75724 Paris Cedex 15, France
| | - Bernard Payrastre
- INSERM U1048, I2MC and Université Paul Sabatier, 31432 Toulouse, France; CHU de Toulouse, Laboratoire d'Hématologie, 31059 Toulouse Cedex 03, France
| | - Hélène Tronchère
- INSERM U1048, I2MC and Université Paul Sabatier, 31432 Toulouse, France.
| |
Collapse
|
47
|
Rappaport J, Manthe RL, Solomon M, Garnacho C, Muro S. A Comparative Study on the Alterations of Endocytic Pathways in Multiple Lysosomal Storage Disorders. Mol Pharm 2016; 13:357-368. [PMID: 26702793 DOI: 10.1021/acs.molpharmaceut.5b00542] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Many cellular activities and pharmaceutical interventions involve endocytosis and delivery to lysosomes for processing. Hence, lysosomal processing defects can cause cell and tissue damage, as in lysosomal storage diseases (LSDs) characterized by lysosomal accumulation of undegraded materials. This storage causes endocytic and trafficking alterations, which exacerbate disease and hinder treatment. However, there have been no systematic studies comparing different endocytic routes in LSDs. Here, we used genetic and pharmacological models of four LSDs (type A Niemann-Pick, type C Niemann-Pick, Fabry, and Gaucher diseases) and evaluated the pinocytic and receptor-mediated activity of the clathrin-, caveolae-, and macropinocytic routes. Bulk pinocytosis was diminished in all diseases, suggesting a generic endocytic alteration linked to lysosomal storage. Fluid-phase (dextran) and ligand (transferrin) uptake via the clathrin route were lower for all LSDs. Fluid-phase and ligand (cholera toxin B) uptake via the caveolar route were both affected but less acutely in Fabry or Gaucher diseases. Epidermal growth factor-induced macropinocytosis was altered in Niemann-Pick cells but not other LSDs. Intracellular trafficking of ligands was also distorted in LSD versus wild-type cells. The extent of these endocytic alterations paralleled the level of cholesterol storage in disease cell lines. Confirming this, pharmacological induction of cholesterol storage in wild-type cells disrupted endocytosis, and model therapeutics restored uptake in proportion to their efficacy in attenuating storage. This suggests a proportional and reversible relationship between endocytosis and lipid (cholesterol) storage. By analogy, the accumulation of biological material in other diseases, or foreign material from drugs or their carriers, may cause similar deficits, warranting further investigation.
Collapse
Affiliation(s)
- Jeff Rappaport
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA
| | - Rachel L Manthe
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA
| | - Melani Solomon
- Institute for Bioscience and Biotechnology Research, University of Maryland, 5115 Plant Sciences Building, College Park, MD 20742-4450, USA
| | - Carmen Garnacho
- Department of Normal and Pathological Histology and Cytology, University of Seville School of Medicine, Seville 41009, Spain
| | - Silvia Muro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA.,Institute for Bioscience and Biotechnology Research, University of Maryland, 5115 Plant Sciences Building, College Park, MD 20742-4450, USA
| |
Collapse
|
48
|
Hamill KM, Wexselblatt E, Tong W, Esko JD, Tor Y. Delivery of an active lysosomal enzyme using GNeosomes. J Mater Chem B 2016; 4:5794-5797. [DOI: 10.1039/c6tb01387b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Guanidinoneomycin derivatives incorporated into liposomes were shown to improve delivery of a fluorescent dye and deliver therapeutic amounts of a lysosomal enzyme.
Collapse
Affiliation(s)
- Kristina M. Hamill
- Department of Chemistry and Biochemistry
- University of California, San Diego
- La Jolla
- USA
| | - Ezequiel Wexselblatt
- Department of Chemistry and Biochemistry
- University of California, San Diego
- La Jolla
- USA
| | - Wenyong Tong
- Cellular and Molecular Medicine
- University of California, San Diego
- La Jolla
- USA
| | - Jeffrey D. Esko
- Cellular and Molecular Medicine
- University of California, San Diego
- La Jolla
- USA
| | - Yitzhak Tor
- Department of Chemistry and Biochemistry
- University of California, San Diego
- La Jolla
- USA
| |
Collapse
|
49
|
Shuvaev VV, Brenner JS, Muzykantov VR. Targeted endothelial nanomedicine for common acute pathological conditions. J Control Release 2015; 219:576-595. [PMID: 26435455 DOI: 10.1016/j.jconrel.2015.09.055] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/24/2015] [Accepted: 09/25/2015] [Indexed: 12/16/2022]
Abstract
Endothelium, a thin monolayer of specialized cells lining the lumen of blood vessels is the key regulatory interface between blood and tissues. Endothelial abnormalities are implicated in many diseases, including common acute conditions with high morbidity and mortality lacking therapy, in part because drugs and drug carriers have no natural endothelial affinity. Precise endothelial drug delivery may improve management of these conditions. Using ligands of molecules exposed to the bloodstream on the endothelial surface enables design of diverse targeted endothelial nanomedicine agents. Target molecules and binding epitopes must be accessible to drug carriers, carriers must be free of harmful effects, and targeting should provide desirable sub-cellular addressing of the drug cargo. The roster of current candidate target molecules for endothelial nanomedicine includes peptidases and other enzymes, cell adhesion molecules and integrins, localized in different domains of the endothelial plasmalemma and differentially distributed throughout the vasculature. Endowing carriers with an affinity to specific endothelial epitopes enables an unprecedented level of precision of control of drug delivery: binding to selected endothelial cell phenotypes, cellular addressing and duration of therapeutic effects. Features of nanocarrier design such as choice of epitope and ligand control delivery and effect of targeted endothelial nanomedicine agents. Pathological factors modulate endothelial targeting and uptake of nanocarriers. Selection of optimal binding sites and design features of nanocarriers are key controllable factors that can be iteratively engineered based on their performance from in vitro to pre-clinical in vivo experimental models. Targeted endothelial nanomedicine agents provide antioxidant, anti-inflammatory and other therapeutic effects unattainable by non-targeted counterparts in animal models of common acute severe human disease conditions. The results of animal studies provide the basis for the challenging translation endothelial nanomedicine into the clinical domain.
Collapse
Affiliation(s)
- Vladimir V Shuvaev
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Center for Translational Targeted Therapeutics and Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Center for Translational Targeted Therapeutics and Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Center for Translational Targeted Therapeutics and Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
50
|
Klingberg H, Loft S, Oddershede LB, Møller P. The influence of flow, shear stress and adhesion molecule targeting on gold nanoparticle uptake in human endothelial cells. NANOSCALE 2015; 7:11409-19. [PMID: 26077188 DOI: 10.1039/c5nr01467k] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The uptake of nanoparticles by endothelial cells is dependent on shear stress adaptation and flow exposure conditions. Adaptation of primary human umbilical vein endothelial cells (HUVECs) to shear stress for 24 h was associated with reduced internalisation of unmodified 80 nm spherical gold nanoparticles (AuNPs) (mean hydrodynamic size of 99 nm in culture medium) after exposure to flow conditions compared with cells that were cultured and exposed to static conditions. Under static conditions, targeting of 80 nm AuNPs conjugated with antibodies against the intracellular adhesion molecule 1 (ICAM-1) (mean hydrodynamic size of 109 nm in culture medium) markedly increased the internalisation of AuNPs in HUVECs that were activated with the tumour necrosis factor (TNF), a treatment that markedly increased the surface expression of ICAM-1. Shear stress-adapted and TNF-activated HUVECs, which were exposed to flow conditions, had higher association with anti-ICAM-1 AuNPs than cells that were not TNF-activated or exposed to particles under static conditions. Hence, shear stress adaptation reduces the uptake of unmodified AuNPs and increases the association between anti-ICAM-1 AuNPs and TNF-activated HUVECs.
Collapse
Affiliation(s)
- Henrik Klingberg
- Department of Public Health, Section of Environment Health, University of Copenhagen, Øster Farimagsgade 5B, DK-1014, Copenhagen, Denmark.
| | | | | | | |
Collapse
|