1
|
Dartora VFC, Passos JS, Costa-Lotufo LV, Lopes LB, Panitch A. Thermosensitive Polymeric Nanoparticles for Drug Co-Encapsulation and Breast Cancer Treatment. Pharmaceutics 2024; 16:231. [PMID: 38399285 PMCID: PMC10892816 DOI: 10.3390/pharmaceutics16020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Despite advances in breast cancer treatment, there remains a need for local management of noninvasive, low-grade ductal carcinoma in situ (DCIS). These focal lesions are well suited for local intraductal treatment. Intraductal administration supported target site drug retention, improved efficacy, and reduced systemic exposure. Here, we used a poly(N-isopropyl acrylamide, pNIPAM) nanoparticle delivery system loaded with cytotoxic piplartine and an MAPKAP Kinase 2 inhibitor (YARA) for this purpose. For tumor environment targeting, a collagen-binding peptide SILY (RRANAALKAGELYKSILYGSG-hydrazide) was attached to pNIPAM nanoparticles, and the nanoparticle diameter, zeta potential, drug loading, and release were assessed. The system was evaluated for cytotoxicity in a 2D cell culture and 3D spheroids. In vivo efficacy was evaluated using a chemical carcinogenesis model in female Sprague-Dawley rats. Nanoparticle delivery significantly reduced the IC50 of piplartine (4.9 times) compared to the drug in solution. The combination of piplartine and YARA in nanoparticles further reduced the piplartine IC50 (~15 times). Treatment with these nanoparticles decreased the in vivo tumor incidence (5.2 times). Notably, the concentration of piplartine in mammary glands treated with nanoparticles (35.3 ± 22.4 μg/mL) was substantially higher than in plasma (0.7 ± 0.05 μg/mL), demonstrating targeted drug retention. These results indicate that our nanocarrier system effectively reduced tumor development with low systemic exposure.
Collapse
Affiliation(s)
- Vanessa Franco Carvalho Dartora
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-900, Brazil; (V.F.C.D.); (J.S.P.); (L.V.C.-L.); (L.B.L.)
- Department of Biomedical Engineering, College of Engineering, University of California Davis, Davis, CA 95616, USA
| | - Julia S. Passos
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-900, Brazil; (V.F.C.D.); (J.S.P.); (L.V.C.-L.); (L.B.L.)
| | - Leticia V. Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-900, Brazil; (V.F.C.D.); (J.S.P.); (L.V.C.-L.); (L.B.L.)
| | - Luciana B. Lopes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-900, Brazil; (V.F.C.D.); (J.S.P.); (L.V.C.-L.); (L.B.L.)
| | - Alyssa Panitch
- Department of Biomedical Engineering, College of Engineering, University of California Davis, Davis, CA 95616, USA
- Wallace H. Coulter Department of Biomedical Engineering, College of Engineering, Georgia Institute of Technology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
2
|
Morelli M, Adcock J, Yim TW, Rook J, Mocco J, Brophy C, Cheung-Flynn J. The Cell Permeant Phosphopetpide mimetic of VASP Alleviates Motor Function Deficits After Experimental Subarachnoid Hemorrhage. J Mol Neurosci 2024; 74:9. [PMID: 38214771 DOI: 10.1007/s12031-023-02180-5] [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: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 01/13/2024]
Abstract
Subarachnoid hemorrhage (SAH) due to the rupture of an intracranial aneurysm leads to delayed vasospasm and neuroischemia, which can result in profound neurologic deficit and death. Therapeutic options after SAH are currently limited to hemodynamic optimization and nimodipine, which have limited clinical efficacy. Experimental SAH results in cerebral vasospasm have demonstrated the downregulation of nitric oxide (NO)-protein kinase G (PKG) signaling elements. VP3 is a novel cell permeant phosphopeptide mimetic of VASP, a substrate of PKG and an actin-associated protein that modulates vasorelaxation in vascular smooth muscle cells. In this study, we determined that intravenous administration of high doses of VP3 did not induce systemic hypotension in rats except at the maximal soluble dose, implying that VP3 is well-tolerated and has a wide therapeutic window. Using a single cisterna magna injection rat model of SAH, we demonstrated that intravenous administration of low-dose VP3 after SAH improved neurologic deficits for up to 14 days as determined by the rotarod test. These findings suggest that strategies aimed at targeting the cerebral vasculature with VP3 may improve neurologic deficits associated with SAH.
Collapse
Affiliation(s)
- Madeleine Morelli
- Department of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jamie Adcock
- Division of Surgical Research, Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tsz Wing Yim
- Department of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jerri Rook
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - J Mocco
- Cerebrovascular Center, Department of Neurosurgery, Mount Sinai Health System, New York, NY, USA
| | - Colleen Brophy
- Department of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joyce Cheung-Flynn
- Department of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
3
|
Dartora VFC, Passos JS, Osorio B, Hung RC, Nguyen M, Wang A, Panitch A. Chitosan hydrogels with MK2 inhibitor peptide-loaded nanoparticles to treat atopic dermatitis. J Control Release 2023; 362:591-605. [PMID: 37660990 DOI: 10.1016/j.jconrel.2023.08.061] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 08/05/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder that lacks ideal long-term treatment options due to a series of side effects, such as skin atrophy, related to the most common treatment prescribed to manage moderate-to-severe AD. In this study, a cell-penetrating MK2 inhibitor peptide YARA (YARAAARQARAKALNRQGLVAA) was loaded into hollow thermo-responsive pNIPAM nanoparticles (NP), which were further incorporated into chitosan hydrogels (H-NP-YARA) to promote local drug delivery, improve moisture and the anti-inflammatory activity. The NPs exhibited high loading efficiency (>50%) and the hydrogel remained porous following NP incorporation as observed by scanning electron microscopy (SEM). Both nanoparticles and hydrogels were able to improve the release of YARA and sustained release to up to 120 h. The hydrogels and NPs delivered 2 and 4-fold more YARA into viable skin layers of porcine skin in vitro at 12 h post-application than the non-encapsulated compound in intact and impaired barrier conditions. Furthermore, the YARA-loaded NPs (NP-YARA) and H-NP-YARA treatment decreased the levels of inflammatory cytokines up to 20 time-fold compared with the non-treated group of human keratinocytes under inflammatory conditions. Consistent with the results in cell culture, the loading of YARA in NP reduced the levels of IL-1β, IL-6, and TNF-α up to 3.3 times in an ex vivo skin culture model after induction of inflammation. A further decrease of up to 17 times-fold was observed with H-NP-YARA treatment compared to the drug in solution. Our data collectively suggest that chitosan hydrogel containing YARA-loaded nanoparticles is a promising new formulation for the topical treatment of AD.
Collapse
Affiliation(s)
- Vanessa F C Dartora
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Julia Sapienza Passos
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Institute of Biomedical Sciences, Department of Pharmacology, University of Sao Paulo, Brazil
| | - Blanca Osorio
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA
| | - Ruei-Chun Hung
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Michael Nguyen
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA
| | - Aijun Wang
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Department of Surgery, University of California Davis, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA
| | - Alyssa Panitch
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Department of Surgery, University of California Davis, Sacramento, CA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA.
| |
Collapse
|
4
|
A comparison of electrosprayed vs vortexed glycosaminoglycan-peptide nanoparticle platform for protection and improved delivery of therapeutic peptides. Colloids Surf B Biointerfaces 2023; 222:113112. [PMID: 36599186 DOI: 10.1016/j.colsurfb.2022.113112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/07/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Therapeutic peptides capable of reducing inflammation via inhibition of the MAP kinase 2 pathway have the potential to reduce inflammation in atopic dermatitis by suppressing secretion of inflammatory cytokines by resident keratinocytes. One of the biggest hurdles to the use of therapeutic peptides, however, is their rapid degradation by intrinsic proteases and peptidases found in serum. Here we introduce a new nanoparticle technology that enhances and extends the bioactivity of a MAP KAP kinase 2 inhibitor peptide (MK2i) via electrostatic complexation with Dermatan sulfate (DS), a glycosaminoglycan, and explore their properties under various conditions. DS-MK2i nanoparticles can be made using electrospray ionization or sonication and vortexing with no stabilizing polymers or crosslinking. Average particle diameter, polydispersity index, and zeta potential were measured over a pH range of 2.5-11.5, in increments of 0.5, in water and at physiological ionic strength. Both particle types were shown to be shelf stable, robust, and behave differently in response to pH. They are also significantly more effective at suppressing cytokine secretion in inflamed, human keratinocytes than peptide alone in the presence of serum, providing a facile method of protecting peptides for therapeutic delivery in conditions such as atopic dermatitis, and abrogating the need for serum-starvation in in vitro testing.
Collapse
|
5
|
Silva S, Kurrikoff K, Langel Ü, Almeida AJ, Vale N. A Second Life for MAP, a Model Amphipathic Peptide. Int J Mol Sci 2022; 23:8322. [PMID: 35955457 PMCID: PMC9368858 DOI: 10.3390/ijms23158322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/26/2022] Open
Abstract
Cell-penetrating peptides (CPP) have been shown to be efficient in the transport of cargoes into the cells, namely siRNA and DNA, proteins and peptides, and in some cases, small therapeutics. These peptides have emerged as a solution to increase drug concentrations in different tissues and various cell types, therefore having a relevant therapeutic relevance which led to clinical trials. One of them, MAP, is a model amphipathic peptide with an α-helical conformation and both hydrophilic and hydrophobic residues in opposite sides of the helix. It is composed of a mixture of alanines, leucines, and lysines (KLALKLALKALKAALKLA). The CPP MAP has the ability to translocate oligonucleotides, peptides and small proteins. However, taking advantage of its unique properties, in recent years innovative concepts were developed, such as in silico studies of modelling with receptors, coupling and repurposing drugs in the central nervous system and oncology, or involving the construction of dual-drug delivery systems using nanoparticles. In addition to designs of MAP-linked vehicles and strategies to achieve highly effective yet less toxic chemotherapy, this review will be focused on unique molecular structure and how it determines its cellular activity, and also intends to address the most recent and frankly motivating issues for the future.
Collapse
Affiliation(s)
- Sara Silva
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal;
| | - Kaido Kurrikoff
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia; (K.K.); (Ü.L.)
| | - Ülo Langel
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia; (K.K.); (Ü.L.)
- Department of Biochemistry and Biophysics, Stockholm University, 10691 Stockholm, Sweden
| | - António J. Almeida
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal;
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
6
|
Tierney JW, Evans BC, Cheung-Flynn J, Wang B, Colazo JM, Polcz ME, Cook RS, Brophy CM, Duvall CL. Therapeutic MK2 inhibition blocks pathological vascular smooth muscle cell phenotype switch. JCI Insight 2021; 6:e142339. [PMID: 34622803 PMCID: PMC8525639 DOI: 10.1172/jci.insight.142339] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Vascular procedures, such as stenting, angioplasty, and bypass grafting, often fail due to intimal hyperplasia (IH), wherein contractile vascular smooth muscle cells (VSMCs) dedifferentiate to synthetic VSMCs, which are highly proliferative, migratory, and fibrotic. Previous studies suggest MAPK-activated protein kinase 2 (MK2) inhibition may limit VSMC proliferation and IH, although the molecular mechanism underlying the observation remains unclear. We demonstrated here that MK2 inhibition blocked the molecular program of contractile to synthetic dedifferentiation and mitigated IH development. Molecular markers of the VSMC contractile phenotype were sustained over time in culture in rat primary VSMCs treated with potent, long-lasting MK2 inhibitory peptide nanopolyplexes (MK2i-NPs), a result supported in human saphenous vein specimens cultured ex vivo. RNA-Seq of MK2i-NP-treated primary human VSMCs revealed programmatic switching toward a contractile VSMC gene expression profile, increasing expression of antiinflammatory and contractile-associated genes while lowering expression of proinflammatory, promigratory, and synthetic phenotype-associated genes. Finally, these results were confirmed using an in vivo rabbit vein graft model where brief, intraoperative treatment with MK2i-NPs decreased IH and synthetic phenotype markers while preserving contractile proteins. These results support further development of MK2i-NPs as a therapy for blocking VSMC phenotype switch and IH associated with cardiovascular procedures.
Collapse
Affiliation(s)
- J. William Tierney
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Brian C. Evans
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of General Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bo Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Monica E. Polcz
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Department of General Surgery and
| | - Rebecca S. Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Colleen M. Brophy
- Division of Vascular Surgery, Department of General Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
7
|
Stiltner J, McCandless K, Zahid M. Cell-Penetrating Peptides: Applications in Tumor Diagnosis and Therapeutics. Pharmaceutics 2021; 13:pharmaceutics13060890. [PMID: 34204007 PMCID: PMC8232808 DOI: 10.3390/pharmaceutics13060890] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 01/27/2023] Open
Abstract
Since their identification over twenty-five years ago, the plethora of cell-penetrating peptides (CPP) and their applications has skyrocketed. These 5 to 30 amino acid in length peptides have the unique property of breaching the cell membrane barrier while carrying cargoes larger than themselves into cells in an intact, functional form. CPPs can be conjugated to fluorophores, activatable probes, radioisotopes or contrast agents for imaging tissues, such as tumors. There is no singular mechanism for translocation of CPPs into a cell, and therefore, many CPPs are taken up by a multitude of cell types, creating the challenge of tumor-specific translocation and hindering clinical effectiveness. Varying strategies have been developed to combat this issue and enhance their diagnostic potential by derivatizing CPPs for better targeting by constructing specific cell-activated forms. These methods are currently being used to image integrin-expressing tumors, breast cancer cells, human histiocytic lymphoma and protease-secreting fibrosarcoma cells, to name a few. Additionally, identifying safe, effective therapeutics for malignant tumors has long been an active area of research. CPPs can circumvent many of the complications found in treating cancer with conventional therapeutics by targeted delivery of drugs into tumors, thereby decreasing off-target side effects, a feat not achievable by currently employed conventional chemotherapeutics. Myriad types of chemotherapeutics such as tyrosine kinase inhibitors, antitumor antibodies and nanoparticles can be functionally attached to these peptides, leading to the possibility of delivering established and novel cancer therapeutics directly to tumor tissue. While much research is needed to overcome potential issues with these peptides, they offer a significant advancement over current mechanisms to treat cancer. In this review, we present a brief overview of the research, leading to identification of CPPs with a comprehensive state-of-the-art review on the role of these novel peptides in both cancer diagnostics as well as therapeutics.
Collapse
Affiliation(s)
| | | | - Maliha Zahid
- Correspondence: ; Tel.: +1-412-692-8893; Fax: 412-692-6184
| |
Collapse
|
8
|
Shafiq M, Jagavelu K, Iqbal H, Yadav P, Chanda D, Verma NK, Ghosh JK, Gaestel M, Hanif K. Inhibition of Mitogen-Activated Protein Kinase (MAPK)-Activated Protein Kinase 2 (MK2) is Protective in Pulmonary Hypertension. Hypertension 2021; 77:1248-1259. [PMID: 33641361 DOI: 10.1161/hypertensionaha.120.15229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Mohammad Shafiq
- From the Division of Pharmacology (M.S., K.J., K.H.), CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research, Ghaziabad, India (M.S., K.J., K.H.)
| | - Kumaravelu Jagavelu
- From the Division of Pharmacology (M.S., K.J., K.H.), CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research, Ghaziabad, India (M.S., K.J., K.H.)
| | - Hina Iqbal
- Department of Molecular Bio-Prospection, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India (H.I., P.Y., D.C.)
| | - Pankaj Yadav
- Department of Molecular Bio-Prospection, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India (H.I., P.Y., D.C.)
| | - Debabrata Chanda
- Department of Molecular Bio-Prospection, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India (H.I., P.Y., D.C.)
| | - Neeraj Kumar Verma
- Division of Molecular and Structural Biology (N.K.V., J.K.G.), CSIR-Central Drug Research Institute, Lucknow, India
| | - Jimut Kanti Ghosh
- Division of Molecular and Structural Biology (N.K.V., J.K.G.), CSIR-Central Drug Research Institute, Lucknow, India
| | - Matthias Gaestel
- Institute for Zellbiochemie, Medizinische Hochschule Hannover (MHH), Hanover, Germany (M.G.)
| | - Kashif Hanif
- From the Division of Pharmacology (M.S., K.J., K.H.), CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research, Ghaziabad, India (M.S., K.J., K.H.)
| |
Collapse
|
9
|
Dailing EA, Kilchrist KV, Tierney JW, Fletcher RB, Evans BC, Duvall CL. Modifying Cell Membranes with Anionic Polymer Amphiphiles Potentiates Intracellular Delivery of Cationic Peptides. ACS APPLIED MATERIALS & INTERFACES 2020; 12:50222-50235. [PMID: 33124813 PMCID: PMC9082340 DOI: 10.1021/acsami.0c13304] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Rapid, facile, and noncovalent cell membrane modification with alkyl-grafted anionic polymers was sought as an approach to enhance intracellular delivery and bioactivity of cationic peptides. We synthesized a library of acrylic acid-based copolymers containing varying amounts of an amine-reactive pentafluorophenyl acrylate monomer followed by postpolymerization modification with a series of alkyl amines to afford precise control over the length and density of aliphatic alkyl side chains. This synthetic strategy enabled systematic investigation of the effect of the polymer structure on membrane binding, potentiation of peptide cell uptake, pH-dependent disruption of lipid bilayers for endosome escape, and intracellular bioavailability. A subset of these polymers exhibited pKa of ∼6.8, which facilitated stable membrane association at physiological pH and rapid, pH-dependent endosomal disruption upon endocytosis as quantified in Galectin-8-YFP reporter cells. Cationic cell penetrating peptide (CPP) uptake was enhanced up to 15-fold in vascular smooth muscle cells in vitro when peptide treatment was preceded by a 30-min pretreatment with lead candidate polymers. We also designed and implemented a new and highly sensitive assay for measuring the intracellular bioavailability of CPPs based on the NanoLuciferase (NanoLuc) technology previously developed for measuring intracellular protein-protein interactions. Using this split luciferase class of assay, polymer pretreatment enhanced intracellular delivery of the CPP-modified HiBiT peptide up to 30-fold relative to CPP-HiBiT without polymer pretreatment (p < 0.05). The overall structural analyses show that polymers containing 50:50 or 70:30 molar ratios of carboxyl groups to alkyl side chains of 6-8 carbons maximized peptide uptake, pH-dependent membrane disruption, and intracellular bioavailability and that this potentiation effect was maximized by pairing with CPPs with high cationic charge density. These results demonstrate a rapid, mild method for polymer modification of cell surfaces to potentiate intracellular delivery, endosome escape, and bioactivity of cationic peptides.
Collapse
Affiliation(s)
- Eric A Dailing
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| | - Kameron V Kilchrist
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| | - J William Tierney
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| | - R Brock Fletcher
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| | - Brian C Evans
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| |
Collapse
|
10
|
Deloney M, Smart K, Christiansen BA, Panitch A. Thermoresponsive, hollow, degradable core-shell nanoparticles for intra-articular delivery of anti-inflammatory peptide. J Control Release 2020; 323:47-58. [PMID: 32278830 PMCID: PMC9930616 DOI: 10.1016/j.jconrel.2020.04.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/13/2022]
Abstract
Inflammation following joint trauma contributes to cartilage degradation and progression of post traumatic osteoarthritis (PTOA). Therefore, drug delivery vehicles that deliver effective anti-inflammatory treatments have the potential to prevent PTOA. We have developed solid and hollow, thermoresponsive nanoparticles for the controlled release of our anti-inflammatory MK2-inhibiting (MK2i) peptide for intra-articular injection to halt inflammation that contributes to the advancement of PTOA. This system exploits the thermosensitive characteristic of N-isopropyl acrylamide (NIPAm) to transition phases when passing through its lower critical solution temperature (LCST). The nanoparticles (NPs) swell below the LCST and constrict above it. Non-crosslinked poly(NIPAm) (pNIPAm), held above its LCST, formed hydrophobic cores around which shells composed of NIPAm, degradable crosslinker N, N'-bis (acryloyl) cystamine (BAC), sulfated 2-acrylamido-2-methyl-1-propanesulfonic acid (AMPS), and acrylic acid (AAc) were polymerized. Removal of the non-crosslinked pNIPAm cores via diffusion produced thermosensitive, degradable nanoparticles with low density, or hollow, cores. The data presented here revealed low-density, termed hollow, nanoparticles (hNPs) load and release significantly more MK2i than solid nanoparticles (sNPs). Furthermore, drug loading below the LCST of NIPAm results in roughly 2.5 times more therapeutic encapsulation compared to loading particles in their constricted state. Hollow nanoparticles increase drug loading compared to solid nanoparticles, are taken up into chondrocytes within 24 h, cleared from the cells within 6 days, significantly decrease the secretion of the proinflammatory cytokine IL-6, and, via intra-articular injection, are successfully delivered into the joint space of rats. The peptide loaded nanoparticles provide a reproducible platform for intra-articular delivery of therapeutics.
Collapse
Affiliation(s)
- Marcus Deloney
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA
| | - Kyra Smart
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA
| | - Blaine A Christiansen
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Department of Orthopedic Surgery, University of California Davis Health, Lawrence J. Ellison Musculoskeletal Research Center, 4635 2nd Avenue, Suite 2000, Sacramento, CA 95817, USA
| | - Alyssa Panitch
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA.
| |
Collapse
|
11
|
Yim TW, Perling D, Polcz M, Komalavilas P, Brophy C, Cheung-Flynn J. A cell permeant phosphopeptide mimetic of Niban inhibits p38 MAPK and restores endothelial function after injury. FASEB J 2020; 34:9180-9191. [PMID: 32396246 PMCID: PMC7383822 DOI: 10.1096/fj.201902745r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022]
Abstract
Vascular injury leads to membrane disruption, ATP release, and endothelial dysfunction. Increases in the phosphorylation of p38 mitogen‐activated protein kinase (p38 MAPK) and decreases in the phosphorylation of Niban, a protein implicated in ER stress and apoptosis, are associated with vascular injury. A cell permeant phosphopeptide mimetic of Niban (NiPp) was generated. The effects of NiPp in restoring endothelial function were determined ex vivo using intact rat aortic tissue (RA) after pharmacological activation of p38 MAPK and also in multiple clinically relevant injury models. Anisomycin (Aniso) increased p38 MAPK phosphorylation and reduced endothelial‐dependent relaxation in RA. Treatment with NiPp prevented Ansio‐induced reduction in endothelial function and increases in p38 MAPK phosphorylation. NiPp treatment also restored endothelial function after stretch injury (subfailure stretch), treatment with acidic Normal Saline (NS), and P2X7R activation with 2′(3′)‐O‐(4‐Benzoylbenzoyl)adenosine 5′‐triphosphate (BzATP). Aged, diseased, human saphenous vein (HSV) remnants obtained from patients undergoing coronary bypass surgical procedures have impaired endothelial function. Treatment of these HSV segments with NiPp improved endothelial‐dependent relaxation. Kinome screening experiments indicated that NiPp inhibits p38 MAPK. These data demonstrate that p38 MAPK and Niban signaling have a role in endothelial function, particularly in response to injury. Niban may represent an endogenous regulator of p38 MAPK activation. The NiPp peptide may serve as an experimental tool to further elucidate p38 MAPK regulation and as a potential therapeutic for endothelial dysfunction.
Collapse
Affiliation(s)
- Tsz Wing Yim
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Daniel Perling
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Monica Polcz
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Padmini Komalavilas
- Department of Surgery, Vanderbilt University, Nashville, TN, USA.,VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Colleen Brophy
- Department of Surgery, Vanderbilt University, Nashville, TN, USA.,VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | | |
Collapse
|
12
|
Brown DI, Cooley BC, Quintana MT, Lander C, Willis MS. Nebulized Delivery of the MAPKAP Kinase 2 Peptide Inhibitor MMI-0100 Protects Against Ischemia-Induced Systolic Dysfunction. Int J Pept Res Ther 2016; 22:317-324. [PMID: 28989333 DOI: 10.1007/s10989-015-9507-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Acute myocardial infarction (AMI) results in systolic dysfunction, myocarditis and fibrotic remodeling, which causes irreversible pathological remodeling of the heart. Associated cell death and inflammation cause cytokine release, which activates the p38 MAPK signaling pathway to propagate damaging signals via MAPKAP kinase 2 (MK2). Previously we showed that intraperitoneal injection of a cell permeable peptide inhibitor of MK2, MMI-0100, protects against fibrosis, apoptosis and systolic dysfunction in a mouse model of AMI induced by left-anterior descending coronary artery (LAD) ligation. Here we tested a new route of administration of MMI-0100: inhalation of nebulized peptide. When given within 30 min of AMI and daily for 2 weeks thereafter, both inhaled and injected MMI-0100 improved cardiac function as measured by conscious echocardiography. Limited fibrosis was observed after 2 weeks by Massons trichrome staining, suggesting that MMI-0100 protects the heart prior to the formation of significant fibrosis. These results support a nebulized route of administration of MMI-0100 can protect the myocardium from ischemic damage.
Collapse
Affiliation(s)
- David I Brown
- McAllister Heart Institute, University of North Carolina, MBRB #2340B, 111 Mason Farm Rd., Chapel Hill, NC, USA
| | - Brian C Cooley
- McAllister Heart Institute, University of North Carolina, MBRB #2340B, 111 Mason Farm Rd., Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, MBRB #2340B, 111 Mason Farm Rd., Chapel Hill, NC, USA
| | - Megan T Quintana
- Department of Surgery, University of North Carolina, MBRB #2340B, 111 Mason Farm Rd., Chapel Hill, NC, USA
| | - Cynthia Lander
- Moerae Matrix, 55 Madison Avenue Suite 400, Morristown, NJ 07960, USA
| | - Monte S Willis
- McAllister Heart Institute, University of North Carolina, MBRB #2340B, 111 Mason Farm Rd., Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, MBRB #2340B, 111 Mason Farm Rd., Chapel Hill, NC, USA.,Department of Pharmacology, University of North Carolina, MBRB #2340B, 111 Mason Farm Rd., Chapel Hill, NC, USA
| |
Collapse
|
13
|
Fiore M, Forli S, Manetti F. Targeting Mitogen-Activated Protein Kinase-Activated Protein Kinase 2 (MAPKAPK2, MK2): Medicinal Chemistry Efforts To Lead Small Molecule Inhibitors to Clinical Trials. J Med Chem 2015; 59:3609-34. [PMID: 26502061 DOI: 10.1021/acs.jmedchem.5b01457] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The p38/MAPK-activated kinase 2 (MK2) pathway is involved in a series of pathological conditions (inflammation diseases and metastasis) and in the resistance mechanism to antitumor agents. None of the p38 inhibitors entered advanced clinical trials because of their unwanted systemic side effects. For this reason, MK2 was identified as an alternative target to block the pathway but avoiding the side effects of p38 inhibition. However, ATP-competitive MK2 inhibitors suffered from low solubility, poor cell permeability, and scarce kinase selectivity. Fortunately, non-ATP-competitive inhibitors of MK2 have been already discovered that allowed circumventing the selectivity issue. These compounds showed the additional advantage to be effective at lower concentrations in comparison to the ATP-competitive inhibitors. Therefore, although the significant difficulties encountered during the development of these inhibitors, MK2 is still considered as an attractive target to treat inflammation and related diseases to prevent tumor metastasis and to increase tumor sensitivity to chemotherapeutics.
Collapse
Affiliation(s)
- Mario Fiore
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena , via A. Moro 2, I-53100 Siena, Italy
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Fabrizio Manetti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena , via A. Moro 2, I-53100 Siena, Italy
| |
Collapse
|
14
|
Evans BC, Hocking KM, Kilchrist KV, Wise ES, Brophy CM, Duvall CL. Endosomolytic Nano-Polyplex Platform Technology for Cytosolic Peptide Delivery To Inhibit Pathological Vasoconstriction. ACS NANO 2015; 9:5893-907. [PMID: 26004140 PMCID: PMC4482421 DOI: 10.1021/acsnano.5b00491] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/24/2015] [Indexed: 05/23/2023]
Abstract
A platform technology has been developed and tested for delivery of intracellular-acting peptides through electrostatically complexed nanoparticles, or nano-polyplexes, formulated from an anionic endosomolytic polymer and cationic therapeutic peptides. This delivery platform has been initially tested and optimized for delivery of two unique vasoactive peptides, a phosphomimetic of heat shock protein 20 and an inhibitor of MAPKAP kinase II, to prevent pathological vasoconstriction (i.e., vasospasm) in human vascular tissue. These peptides inhibit vasoconstriction and promote vasorelaxation by modulating actin dynamics in vascular smooth muscle cells. Formulating these peptides into nano-polyplexes significantly enhances peptide uptake and retention, facilitates cytosolic delivery through a pH-dependent endosomal escape mechanism, and enhances peptide bioactivity in vitro as measured by inhibition of F-actin stress fiber formation. In comparison to treatment with the free peptides, which were endowed with cell-penetrating sequences, the nano-polyplexes significantly increased vasorelaxation, inhibited vasoconstriction, and decreased F-actin formation in the human saphenous vein ex vivo. These results suggest that these formulations have significant potential for treatment of conditions such as cerebral vasospasm following subarachnoid hemorrhage. Furthermore, because many therapeutic peptides include cationic cell-penetrating segments, this simple and modular platform technology may have broad applicability as a cost-effective approach for enhancing the efficacy of cytosolically active peptides.
Collapse
Affiliation(s)
- Brian C. Evans
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB 351826, Nashville, Tennessee 37235, United States
| | - Kyle M. Hocking
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB 351826, Nashville, Tennessee 37235, United States
| | - Kameron V. Kilchrist
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB 351826, Nashville, Tennessee 37235, United States
| | - Eric S. Wise
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, D-5237 Medical Center North, 1161 22nd Avenue South, Nashville, Tennessee 37232, United States
| | - Colleen M. Brophy
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, D-5237 Medical Center North, 1161 22nd Avenue South, Nashville, Tennessee 37232, United States
- Veterans Affairs Medical Center, VA Tennessee Valley Healthcare System, 1310 24th Avenue South, Nashville, Tennessee 37212, United States
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB 351826, Nashville, Tennessee 37235, United States
| |
Collapse
|
15
|
Evans BC, Hocking KM, Osgood MJ, Voskresensky I, Dmowska J, Kilchrist KV, Brophy CM, Duvall CL. MK2 inhibitory peptide delivered in nanopolyplexes prevents vascular graft intimal hyperplasia. Sci Transl Med 2015; 7:291ra95. [PMID: 26062847 PMCID: PMC5371354 DOI: 10.1126/scitranslmed.aaa4549] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Autologous vein grafts are commonly used for coronary and peripheral artery bypass but have a high incidence of intimal hyperplasia (IH) and failure. We present a nanopolyplex (NP) approach that efficiently delivers a mitogen-activated protein kinase (MAPK)-activated protein (MAPKAP) kinase 2 inhibitory peptide (MK2i) to graft tissue to improve long-term patency by inhibiting pathways that initiate IH. In vitro testing in human vascular smooth muscle cells revealed that formulation into MK2i-NPs increased cell internalization, endosomal escape, and intracellular half-life of MK2i. This efficient delivery mechanism enabled MK2i-NPs to sustain potent inhibition of inflammatory cytokine production and migration in vascular cells. In intact human saphenous vein, MK2i-NPs blocked inflammatory and migratory signaling, as confirmed by reduced phosphorylation of the posttranscriptional gene regulator heterogeneous nuclear ribonucleoprotein A0, the transcription factor cAMP (adenosine 3',5'-monophosphate) element-binding protein, and the chaperone heat shock protein 27. The molecular effects of MK2i-NPs caused functional inhibition of IH in human saphenous vein cultured ex vivo. In a rabbit vein transplant model, a 30-min intraoperative graft treatment with MK2i-NPs significantly reduced in vivo IH 28 days posttransplant compared with untreated or free MK2i-treated grafts. The decrease in IH in MK2i-NP-treated grafts in the rabbit model also corresponded with decreased cellular proliferation and maintenance of the vascular wall smooth muscle cells in a more contractile phenotype. These data indicate that nanoformulated MK2 inhibitors are a promising strategy for preventing graft failure.
Collapse
Affiliation(s)
- Brian C Evans
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kyle M Hocking
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Michael J Osgood
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Igor Voskresensky
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Julia Dmowska
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kameron V Kilchrist
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Colleen M Brophy
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA. Veterans Affairs Medical Center, VA Tennessee Valley Healthcare System, Nashville TN 37212, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
16
|
Brugnano J, Ward BC, Panitch A. Cell penetrating peptides can exert biological activity: a review. Biomol Concepts 2015; 1:109-16. [PMID: 25961990 DOI: 10.1515/bmc.2010.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Cell penetrating peptides (CPPs) have been successful in delivering cargo into many different cell types and are an important alternative to other methods of permeation that might damage the integrity of the cell membrane. The traditional view of CPPs is that they are inert molecules that can be successfully used to deliver many cargos intracellularly. The goal of this review is to challenge this traditional understanding of CPPs. Recent literature has demonstrated that CPPs themselves can convey biological activity, including the alteration of gene expression and inhibition of protein kinases and proteolytic activity. Further characterization of CPPs is required to determine the extent of this activity. Research into the use of CPPs for intracellular delivery should continue with investigators being aware of these recent results.
Collapse
|
17
|
Moutal A, François-Moutal L, Brittain JM, Khanna M, Khanna R. Differential neuroprotective potential of CRMP2 peptide aptamers conjugated to cationic, hydrophobic, and amphipathic cell penetrating peptides. Front Cell Neurosci 2015; 8:471. [PMID: 25674050 PMCID: PMC4306314 DOI: 10.3389/fncel.2014.00471] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 12/30/2014] [Indexed: 01/26/2023] Open
Abstract
The microtubule-associated axonal specification collapsin response mediator protein 2 (CRMP2) is a novel target for neuroprotection. A CRMP2 peptide (TAT-CBD3) conjugated to the HIV transactivator of transcription (TAT) protein's cationic cell penetrating peptide (CPP) motif protected neurons in the face of toxic levels of Ca(2+) influx leaked in via N-methyl-D-aspartate receptor (NMDAR) hyperactivation. Here we tested whether replacing the hydrophilic TAT motif with alternative cationic (nona-arginine (R9)), hydrophobic (membrane transport sequence (MTS) of k-fibroblast growth factor) or amphipathic (model amphipathic peptide (MAP)) CPPs could be superior to the neuroprotection bestowed by TAT-CBD3. In giant plasma membrane vesicles (GPMVs) derived from cortical neurons, the peptides translocated across plasma membranes with similar efficiencies. Cortical neurons, acutely treated with peptides prior to a toxic glutamate challenge, demonstrated enhanced efflux of R9-CBD3 compared to others. R9-CBD3 inhibited N-methyl-D-aspartate (NMDA)-evoked Ca(2+) influx to a similar extent as TAT-CBD3 while MTS-CBD3 was ineffective which correlated with the ability of R9- and TAT-CBD3, but not MTS-CBD3, to block NMDAR interaction with CRMP2. Unrestricted Ca(2+) influx through NMDARs leading to delayed calcium dysregulation and neuronal cell death was blocked by all peptides but MAP-CBD3. When applied acutely for 10 min, R9-CBD3 was more effective than TAT-CBD3 at neuroprotection while MTS- and MAP-CBD3 were ineffective. In contrast, long-term (>24 h) treatment with MTS-CBD3 conferred neuroprotection where TAT-CBD3 failed. Neither peptide altered surface trafficking of NMDARs. Neuroprotection conferred by MTS-CBD3 peptide is likely due to its increased uptake coupled with decreased efflux when compared to TAT-CBD3. Overall, our results demonstrate that altering CPPs can bestow differential neuroprotective potential onto the CBD3 cargo.
Collapse
Affiliation(s)
- Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona Tucson, AZ, USA
| | | | - Joel M Brittain
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine Indianapolis, IN, USA
| | - May Khanna
- Department of Pharmacology, College of Medicine, University of Arizona Tucson, AZ, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona Tucson, AZ, USA ; Neuroscience Graduate Interdisciplinary Program, College of Medicine, University of Arizona Tucson, AZ, USA
| |
Collapse
|
18
|
Mohamed MF, Hammac GK, Guptill L, Seleem MN. Antibacterial activity of novel cationic peptides against clinical isolates of multi-drug resistant Staphylococcus pseudintermedius from infected dogs. PLoS One 2014; 9:e116259. [PMID: 25551573 PMCID: PMC4281220 DOI: 10.1371/journal.pone.0116259] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 12/04/2014] [Indexed: 11/18/2022] Open
Abstract
Staphylococcus pseudintermedius is a major cause of skin and soft tissue infections in companion animals and has zoonotic potential. Additionally, methicillin-resistant S. pseudintermedius (MRSP) has emerged with resistance to virtually all classes of antimicrobials. Thus, novel treatment options with new modes of action are required. Here, we investigated the antimicrobial activity of six synthetic short peptides against clinical isolates of methicillin-susceptible and MRSP isolated from infected dogs. All six peptides demonstrated potent anti-staphylococcal activity regardless of existing resistance phenotype. The most effective peptides were RRIKA (with modified C terminus to increase amphipathicity and hydrophobicity) and WR-12 (α-helical peptide consisting exclusively of arginine and tryptophan) with minimum inhibitory concentration50 (MIC50) of 1 µM and MIC90 of 2 µM. RR (short anti-inflammatory peptide) and IK8 “D isoform” demonstrated good antimicrobial activity with MIC50 of 4 µM and MIC90 of 8 µM. Penetratin and (KFF)3K (two cell penetrating peptides) were the least effective with MIC50 of 8 µM and MIC90 of 16 µM. Killing kinetics revealed a major advantage of peptides over conventional antibiotics, demonstrating potent bactericidal activity within minutes. Studies with propidium iodide and transmission electron microscopy revealed that peptides damaged the bacterial membrane leading to leakage of cytoplasmic contents and consequently, cell death. A potent synergistic increase in the antibacterial effect of the cell penetrating peptide (KFF)3K was noticed when combined with other peptides and with antibiotics. In addition, all peptides displayed synergistic interactions when combined together. Furthermore, peptides demonstrated good therapeutic indices with minimal toxicity toward mammalian cells. Resistance to peptides did not evolve after 10 passages of S. pseudintermedius at sub-inhibitory concentration. However, the MICs of amikacin and ciprofloxacin increased 32 and 8 fold, respectively; under similar conditions. Taken together, these results support designing of peptide-based therapeutics for combating MRSP infections, particularly for topical application.
Collapse
Affiliation(s)
- Mohamed F Mohamed
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
| | - G Kenitra Hammac
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
| | - Lynn Guptill
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
| | - Mohamed N Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
19
|
Mohamed MF, Seleem MN. Efficacy of short novel antimicrobial and anti-inflammatory peptides in a mouse model of methicillin-resistant Staphylococcus aureus (MRSA) skin infection. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:1979-83. [PMID: 25378910 PMCID: PMC4207544 DOI: 10.2147/dddt.s72129] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The therapeutic efficacy of two novel short antimicrobial and anti-inflammatory peptides (RR and RRIKA) was evaluated in a mouse model of staphylococcal skin infection. RR (2%) and RRIKA (2%) significantly reduced the bacterial counts and the levels of proinflammatory cytokines, tumor necrosis factor (TNF)-α, and interleukin (IL)-6, in methicillin-resistant Staphylococcus aureus USA 300-0114 skin lesions. Furthermore, the combined therapy of RRIKA (1%) and lysostaphin (0.5%) had significantly higher antistaphylococcal and anti-inflammatory activity compared to monotherapy. This study supports the potential use of these peptides for topical treatment of methicillin-resistant Staphylococcus aureus skin infections.
Collapse
Affiliation(s)
- Mohamed F Mohamed
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Mohamed N Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
20
|
MMI-0100 inhibits cardiac fibrosis in myocardial infarction by direct actions on cardiomyocytes and fibroblasts via MK2 inhibition. J Mol Cell Cardiol 2014; 77:86-101. [PMID: 25257914 DOI: 10.1016/j.yjmcc.2014.09.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 09/07/2014] [Accepted: 09/10/2014] [Indexed: 12/12/2022]
Abstract
The cell-permeant peptide inhibitor of MAPKAP kinase 2 (MK2), MMI-0100, inhibits MK2 and downstream fibrosis and inflammation. Recent studies have demonstrated that MMI-0100 reduces intimal hyperplasia in a mouse vein graft model, pulmonary fibrosis in a murine bleomycin-induced model and development of adhesions in conjunction with abdominal surgery. MK2 is critical to the pathogenesis of ischemic heart injury as MK2(-/-) mice are resistant to ischemic remodeling. Therefore, we tested the hypothesis that inhibiting MK2 with MMI-0100 would protect the heart after acute myocardial infarction (AMI) in vivo. AMI was induced by placing a permanent LAD coronary ligation. When MMI-0100 peptide was given 30 min after permanent LAD coronary artery ligation, the resulting fibrosis was reduced/prevented ~50% at a 2 week time point, with a corresponding improvement in cardiac function and decrease in left ventricular dilation. In cultured cardiomyocytes and fibroblasts, MMI-0100 inhibited MK2 to reduce cardiomyocyte caspase 3/7 activity, while enhancing primary cardiac fibroblast caspase 3/7 activity, which may explain MMI-0100's salvage of cardiac function and anti-fibrotic effects in vivo. These findings suggest that therapeutic inhibition of MK2 after acute MI, using rationally-designed cell-permeant peptides, inhibits cardiac fibrosis and maintains cardiac function by mechanisms that involve inhibiting cardiomyocyte apoptosis, while enhancing primary cardiac fibroblast cell death.
Collapse
|
21
|
Targeting methicillin-resistant Staphylococcus aureus with short salt-resistant synthetic peptides. Antimicrob Agents Chemother 2014; 58:4113-22. [PMID: 24798285 DOI: 10.1128/aac.02578-14] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The seriousness of microbial resistance combined with the lack of new antimicrobials has increased interest in the development of antimicrobial peptides (AMPs) as novel therapeutics. In this study, we evaluated the antimicrobial activities of two short synthetic peptides, namely, RRIKA and RR. These peptides exhibited potent antimicrobial activity against Staphylococcus aureus, and their antimicrobial effects were significantly enhanced by addition of three amino acids in the C terminus, which consequently increased the amphipathicity, hydrophobicity, and net charge. Moreover, RRIKA and RR demonstrated a significant and rapid bactericidal effect against clinical and drug-resistant Staphylococcus isolates, including methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-intermediate S. aureus (VISA), vancomycin-resistant S. aureus (VRSA), linezolid-resistant S. aureus, and methicillin-resistant Staphylococcus epidermidis. In contrast to many natural AMPs, RRIKA and RR retained their activity in the presence of physiological concentrations of NaCl and MgCl2. Both RRIKA and RR enhanced the killing of lysostaphin more than 1,000-fold and eradicated MRSA and VRSA isolates within 20 min. Furthermore, the peptides presented were superior in reducing adherent biofilms of S. aureus and S. epidermidis compared to results with conventional antibiotics. Our findings indicate that the staphylocidal effects of our peptides were through permeabilization of the bacterial membrane, leading to leakage of cytoplasmic contents and cell death. Furthermore, peptides were not toxic to HeLa cells at 4- to 8-fold their antimicrobial concentrations. The potent and salt-insensitive antimicrobial activities of these peptides present an attractive therapeutic candidate for treatment of multidrug-resistant S. aureus infections.
Collapse
|
22
|
Tints K, Prink M, Neuman T, Palm K. LXXLL peptide converts transportan 10 to a potent inducer of apoptosis in breast cancer cells. Int J Mol Sci 2014; 15:5680-98. [PMID: 24705462 PMCID: PMC4013589 DOI: 10.3390/ijms15045680] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 03/18/2014] [Accepted: 03/24/2014] [Indexed: 12/15/2022] Open
Abstract
Degenerate expression of transcription coregulator proteins is observed in most human cancers. Therefore, in targeted anti-cancer therapy development, intervention at the level of cancer-specific transcription is of high interest. The steroid receptor coactivator-1 (SRC-1) is highly expressed in breast, endometrial, and prostate cancer. It is present in various transcription complexes, including those containing nuclear hormone receptors. We examined the effects of a peptide that contains the LXXLL-motif of the human SRC-1 nuclear receptor box 1 linked to the cell-penetrating transportan 10 (TP10), hereafter referred to as TP10-SRC1LXXLL, on proliferation and estrogen-mediated transcription of breast cancer cells in vitro. Our data show that TP10-SRC1LXXLL induced dose-dependent cell death of breast cancer cells, and that this effect was not affected by estrogen receptor (ER) status. Surprisingly TP10-SRC1LXXLL severely reduced the viability and proliferation of hormone-unresponsive breast cancer MDA-MB-231 cells. In addition, the regulation of the endogenous ERα direct target gene pS2 was not affected by TP10-SRC1LXXLL in estrogen-stimulated MCF-7 cells. Dermal fibroblasts were similarly affected by treatment with higher concentrations of TP10-SRC1LXXLL and this effect was significantly delayed. These results suggest that the TP10-SRC1LXXLL peptide may be an effective drug candidate in the treatment of cancers with minimal therapeutic options, for example ER-negative tumors.
Collapse
Affiliation(s)
- Kairit Tints
- Protobios LLC, Mäealuse 4, Tallinn 12618, Estonia.
| | - Madis Prink
- Protobios LLC, Mäealuse 4, Tallinn 12618, Estonia.
| | | | - Kaia Palm
- Protobios LLC, Mäealuse 4, Tallinn 12618, Estonia.
| |
Collapse
|
23
|
Gaestel M. What goes up must come down: molecular basis of MAPKAP kinase 2/3-dependent regulation of the inflammatory response and its inhibition. Biol Chem 2014; 394:1301-15. [PMID: 23832958 DOI: 10.1515/hsz-2013-0197] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/01/2013] [Indexed: 12/21/2022]
Abstract
Inflammation is normally a fast and transient response to microbial invaders or sterile damage and has to be stringently controlled. The closely-related mitogen-activated protein kinase-activated protein kinases MK2 and MK3 are involved in both up- and down-regulation of inflammation in mammals and govern the inflammatory response at different regulatory levels of gene expression and with different kinetics. In conjunction with their activator MAP kinase p38, MK2 and MK3 stimulate the transcription of immediate-early genes including that of the mRNA-binding protein tristetraprolin (TTP). TTP competes with the constitutively expressed protein human antigen R in binding to the mRNA destabilizing adenylate-uridylate -rich element. MK2 and MK3 also regulate the activity of TTP by direct phosphorylation, determine stability and stimulate the translation of cytokine mRNAs. In addition, TTP controls its own re-synthesis via stability and translation of its mRNA in a phosphorylation-dependent manner. This results in a complex scenario of gene expression and guarantees fast up-regulation and intrinsic feedback control of the inflammatory response of macrophages. Inhibition of MK2/3 by small-molecule pharmaceutical inhibitors is an emerging strategy to manipulate the inflammatory response as a therapeutic option. This strategy could display advantages over the direct inhibition of MAP kinase p38.
Collapse
|
24
|
Copolovici DM, Langel K, Eriste E, Langel Ü. Cell-penetrating peptides: design, synthesis, and applications. ACS NANO 2014; 8:1972-94. [PMID: 24559246 DOI: 10.1021/nn4057269] [Citation(s) in RCA: 694] [Impact Index Per Article: 63.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The intrinsic property of cell-penetrating peptides (CPPs) to deliver therapeutic molecules (nucleic acids, drugs, imaging agents) to cells and tissues in a nontoxic manner has indicated that they may be potential components of future drugs and disease diagnostic agents. These versatile peptides are simple to synthesize, functionalize, and characterize yet are able to deliver covalently or noncovalently conjugated bioactive cargos (from small chemical drugs to large plasmid DNA) inside cells, primarily via endocytosis, in order to obtain high levels of gene expression, gene silencing, or tumor targeting. Typically, CPPs are often passive and nonselective yet must be functionalized or chemically modified to create effective delivery vectors that succeed in targeting specific cells or tissues. Furthermore, the design of clinically effective systemic delivery systems requires the same amount of attention to detail in both design of the delivered cargo and the cell-penetrating peptide used to deliver it.
Collapse
Affiliation(s)
- Dana Maria Copolovici
- Laboratory of Molecular Biotechnology, Institute of Technology, Tartu University , 504 11 Tartu, Estonia
| | | | | | | |
Collapse
|
25
|
Vittal R, Fisher A, Gu H, Mickler EA, Panitch A, Lander C, Cummings OW, Sandusky GE, Wilkes DS. Peptide-mediated inhibition of mitogen-activated protein kinase-activated protein kinase-2 ameliorates bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol 2013; 49:47-57. [PMID: 23470623 DOI: 10.1165/rcmb.2012-0389oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Mitogen-activated protein kinase-activated protein kinase-2 (MAPKAPK2, or MK2), a serine/threonine kinase downstream of p38 mitogen-activated protein kinase, has been implicated in inflammation and fibrosis. Compared with pathologically normal lung tissue, significantly higher concentrations of activated MK2 are evident in lung biopsies of patients with idiopathic pulmonary fibrosis (IPF). Expression is localized to fibroblasts and epithelial cells. In the murine bleomycin model of pulmonary fibrosis, we observed robust, activated MK2 expression on Day 7 (prefibrotic stage) and Day 14 (postfibrotic stage). To determine the effects of MK2 inhibition during the postinflammatory/prefibrotic and postfibrotic stages, C57BL/6 mice received intratracheal bleomycin instillation (0.025 U; Day 0), followed by PBS or the MK2 inhibitor (MK2i; 37.5 μg/kg), administered via either local (nebulized) or systemic (intraperitoneal) routes. MK2i or PBS was dosed daily for 14 days subsequent to bleomycin injury, beginning on either Day 7 or Day 14. Regardless of mode of administration or stage of intervention, MK2i significantly abrogated collagen deposition, myofibroblast differentiation and activated MK2 expression. MK2i also decreased circulating TNF-α and IL-6 concentrations, and modulated the local mRNA expression of profibrotic cytokine il-1β, matrix-related genes col1a2, col3a1, and lox, and transforming growth factor-β family members, including smad3, serpine1 (pai1), and smad6/7. In vitro, MK2i dose-dependently attenuated total MK2, myofibroblast differentiation, the secretion of collagen Type I, fibronectin, and the activation of focal adhesion kinase, whereas activated MK2 was attenuated at optimal doses. The peptide-mediated inhibition of MK2 affects both inflammatory and fibrotic responses, and thus may offer a promising therapeutic target for IPF.
Collapse
Affiliation(s)
- Ragini Vittal
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Brugnano J, McMasters J, Panitch A. Characterization of endocytic uptake of MK2-inhibitor peptides. J Pept Sci 2013; 19:629-38. [PMID: 24014473 DOI: 10.1002/psc.2541] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 06/25/2013] [Accepted: 06/26/2013] [Indexed: 12/31/2022]
Abstract
Cell penetrating peptides (CPP) have been widely used to increase the cellular delivery of their associated cargo. Multiple modes of uptake have been identified; however, they cannot be predicted a priori. Elucidating these mechanisms is important for understanding peptide function as well as further optimizing cellular delivery. We have developed a class of mitogen activated protein kinase activated protein kinase 2 (MK2) inhibitor peptides, named FAK and YARA that utilize CPP domains to gain cellular access. In this study, we investigate the mechanism of endocytosis of these MK2 inhibitors by examining the uptake of fluorescently labeled peptide in human monocyte (THP-1) and mesothelial cells, and looking for colocalization with known markers of endocytosis. Our results indicate that uptake of the MK2 inhibitors was minimally enhanced by the addition of the fluorescent label, and that the type of endocytosis used by the inhibitor depends on several factors including concentration, cell type, and which CPP was used. We found that in THP-1 cells, the uptake of YARA occurred primarily via macropinocytosis, whereas FAK entered via all three mechanisms of endocytosis examined in this study. In mesothelial cells, uptake of YARA occurred via caveolae-mediated endocytosis, but became less specific at higher concentrations; whereas uptake of FAK occurred through clathrin-mediated endocytosis. In all cases, the delivery resulted in active inhibition of MK2. In summary, the results support endocytic uptake of fluorescently labeled FAK and YARA in two different cell lines, with the mechanism of uptake dependent on extracellular concentration, cell type, and choice of CPP.
Collapse
Affiliation(s)
- Jamie Brugnano
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN, 47907, USA
| | | | | |
Collapse
|
27
|
Howl J, Matou-Nasri S, West DC, Farquhar M, Slaninová J, Ostenson CG, Zorko M, Ostlund P, Kumar S, Langel U, McKeating J, Jones S. Bioportide: an emergent concept of bioactive cell-penetrating peptides. Cell Mol Life Sci 2012; 69:2951-66. [PMID: 22527714 PMCID: PMC11114504 DOI: 10.1007/s00018-012-0979-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 03/20/2012] [Accepted: 03/22/2012] [Indexed: 12/22/2022]
Abstract
Cell-penetrating peptides (CPPs) have proven utility for the highly efficient intracellular delivery of bioactive cargoes that include peptides, proteins, and oligonucleotides. The many strategies developed to utilize CPPs solely as pharmacokinetic modifiers necessarily requires them to be relatively inert. Moreover, it is feasible to combine one or multiple CPPs with bioactive cargoes either by direct chemical conjugation or, more rarely, as non-covalent complexes. In terms of the message-address hypothesis, this combination of cargo (message) linked to a CPP (address) as a tandem construct conforms to the sychnological organization. More recently, we have introduced the term bioportide to describe monomeric CPPs that are intrinsically bioactive. Herein, we describe the design and biochemical properties of two rhegnylogically organized monometic CPPs that collectively modulate a variety of biological and pathophysiological phenomena. Thus, camptide, a cell-penetrant sequence located within the first intracellular loop of a human calcitonin receptor, regulates cAMP-dependent processes to modulate insulin secretion and viral infectivity. Nosangiotide, a bioportide derived from endothelial nitric oxide synthase, potently inhibits many aspects of the endothelial cell morphology and movement and displays potent anti-angiogenic activity in vivo. We conclude that, due to their capacity to translocate and target intracellular signaling events, bioportides represent an innovative generic class of bioactive agents.
Collapse
Affiliation(s)
- John Howl
- Research Institute in Healthcare Science, School of Applied Sciences, University of Wolverhampton, Wolverhampton, WV1 1LY, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hu L, Paul Fawcett J, Gu J. Protein target discovery of drug and its reactive intermediate metabolite by using proteomic strategy. Acta Pharm Sin B 2012. [DOI: 10.1016/j.apsb.2012.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
29
|
Bartlett RL, Medow MR, Panitch A, Seal B. Hemocompatible Poly(NIPAm-MBA-AMPS) Colloidal Nanoparticles as Carriers of Anti-inflammatory Cell Penetrating Peptides. Biomacromolecules 2012; 13:1204-11. [PMID: 22452800 DOI: 10.1021/bm300173x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Rush L Bartlett
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive,
West Lafayette, Indiana, 47907
| | - Matthew R. Medow
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive,
West Lafayette, Indiana, 47907
| | - Alyssa Panitch
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive,
West Lafayette, Indiana, 47907
| | - Brandon Seal
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive,
West Lafayette, Indiana, 47907
| |
Collapse
|
30
|
Muto A, Panitch A, Kim N, Park K, Komalavilas P, Brophy CM, Dardik A. Inhibition of Mitogen Activated Protein Kinase Activated Protein Kinase II with MMI-0100 reduces intimal hyperplasia ex vivo and in vivo. Vascul Pharmacol 2011; 56:47-55. [PMID: 22024359 DOI: 10.1016/j.vph.2011.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 05/24/2011] [Accepted: 07/06/2011] [Indexed: 10/16/2022]
Abstract
Vein graft intimal hyperplasia remains the leading cause of graft failure, despite many pharmacological approaches that have failed to translate to human therapy. We investigated whether local suppression of inflammation and fibrosis with MMI-0100, a novel peptide inhibitor of Mitogen Activated Protein Kinase Activated Protein Kinase II (MK2), would be an alternative strategy to reduce cell proliferation and intimal hyperplasia. The cell permeant peptide MMI-0100 was synthesized using standard Fmoc chemistry. Pharmacological doses of MMI-0100 induced minimal human endothelial and smooth muscle cell proliferation (30% and 12% respectively). MMI-0100 suppressed IL-6 expression to control levels, without effect on IL-8 expression. MMI-0100 caused sodium nitroprusside induced smooth muscle cell relaxation and inhibited intimal thickening in human saphenous vein rings in a dose-dependent fashion. In a murine aortic bypass model, MMI-0100 reduced intimal thickness in vein grafts by 72%, and there were fewer F4/80-reactive cells in vein grafts treated with MMI-0100. MMI-0100 prevents vein graft intimal thickening ex vivo and in vivo. These results suggest that inhibition of MK2 with the cell-permeant peptide MMI-0100 may be a novel strategy to suppress fibrotic processes such as vein graft disease.
Collapse
Affiliation(s)
- Akihito Muto
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Woolley AJ, Desai HA, Steckbeck MA, Patel NK, Otto KJ. In situ characterization of the brain-microdevice interface using device-capture histology. J Neurosci Methods 2011; 201:67-77. [PMID: 21802446 PMCID: PMC3179652 DOI: 10.1016/j.jneumeth.2011.07.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 06/21/2011] [Accepted: 07/13/2011] [Indexed: 11/30/2022]
Abstract
Accurate assessment of brain-implantable microdevice bio-integration remains a formidable challenge. Prevailing histological methods require device extraction prior to tissue processing, often disrupting and removing the tissue of interest which had been surrounding the device. The Device-Capture Histology method, presented here, overcomes many limitations of the conventional Device-Explant Histology method, by collecting the device and surrounding tissue intact for subsequent labeling. With the implant remaining in situ, accurate and precise imaging of the morphologically preserved tissue at the brain/microdevice interface can then be collected and quantified. First, this article presents the Device-Capture Histology method for obtaining and processing the intact, undisturbed microdevice-tissue interface, and imaging using fluorescent labeling and confocal microscopy. Second, this article gives examples of how to quantify features found in the captured peridevice tissue. We also share histological data capturing (1) the impact of microdevice implantation on tissue, (2) the effects of an experimental anti-inflammatory coating, (3) a dense grouping of cell nuclei encapsulating a long-term implant, and (4) atypical oligodendrocyte organization neighboring a long term implant. Data sets collected using the Device-Capture Histology method are presented to demonstrate the significant advantages of processing the intact microdevice-tissue interface, and to underscore the utility of the method in understanding the effects of the brain-implantable microdevices on nearby tissue.
Collapse
Affiliation(s)
- Andrew J. Woolley
- Department of Biological Sciences, Purdue University, 915 West State Street, West Lafayette, IN, 47907-2054, United States
| | - Himanshi A. Desai
- Department of Biological Sciences, Purdue University, 915 West State Street, West Lafayette, IN, 47907-2054, United States
| | - Mitchell A. Steckbeck
- Department of Biological Sciences, Purdue University, 915 West State Street, West Lafayette, IN, 47907-2054, United States
| | - Neil K. Patel
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN, 47907-2032, United States
| | - Kevin J. Otto
- Department of Biological Sciences, Purdue University, 915 West State Street, West Lafayette, IN, 47907-2054, United States
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN, 47907-2032, United States
| |
Collapse
|
32
|
Li H, Nelson CE, Evans BC, Duvall CL. Delivery of intracellular-acting biologics in pro-apoptotic therapies. Curr Pharm Des 2011; 17:293-319. [PMID: 21348831 DOI: 10.2174/138161211795049642] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 02/18/2011] [Indexed: 12/21/2022]
Abstract
The recent elucidation of molecular regulators of apoptosis and their roles in cellular oncogenesis has motivated the development of biomacromolecular anticancer therapeutics that can activate intracellular apoptotic signaling pathways. Pharmaceutical scientists have employed a variety of classes of biologics toward this goal, including antisense oligodeoxynucleotides, small interfering RNA, proteins, antibodies, and peptides. However, stability in the in vivo environment, tumor-specific biodistribution, cell internalization, and localization to the intracellular microenvironment where the targeted molecule is localized pose significant challenges that limit the ability to directly apply intracellular-acting, pro-apoptotic biologics for therapeutic use. Thus, approaches to improve the pharmaceutical properties of therapeutic biomacromolecules are of great significance and have included chemically modifying the bioactive molecule itself or formulation with auxiliary compounds. Recently, promising advances in delivery of pro-apoptotic biomacromolecular agents have been made using tools such as peptide "stapling", cell penetrating peptides, fusogenic peptides, liposomes, nanoparticles, smart polymers, and synergistic combinations of these components. This review will discuss the molecular mediators of cellular apoptosis, the respective mechanisms by which these mediators are dysregulated in cellular oncogenesis, the history and development of both nucleic-acid and amino-acid based drugs, and techniques to achieve intracellular delivery of these biologics. Finally, recent applications where pro-apoptotic functionality has been achieved through delivery of intracellular-acting biomacromolecular drugs will be highlighted.
Collapse
Affiliation(s)
- Hongmei Li
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | | | | | | |
Collapse
|
33
|
Brugnano JL, Chan BK, Seal BL, Panitch A. Cell-penetrating peptides can confer biological function: regulation of inflammatory cytokines in human monocytes by MK2 inhibitor peptides. J Control Release 2011; 155:128-33. [PMID: 21600941 DOI: 10.1016/j.jconrel.2011.05.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 05/03/2011] [Indexed: 01/08/2023]
Abstract
Cell-penetrating peptides have been used as a method of delivering biologically active peptide for over two decades. In this paper, we covalently attached four different cell-penetrating peptides to a peptide that inhibits a kinase important in inflammation, mitogen-activated protein kinase activated protein kinase 2 (MAPKAP2 or MK2). We evaluated the specificity, toxicity, and functionality of these therapeutics in an in vitro model of inflammation using THP-1 monocytes. When treated with the MK2 peptide inhibitors, activated THP-1 human monocytes challenged with lipopolysaccharide (LPS) showed a decrease in TNF-α and IL-6 excretion without apparent toxicity. In addition, western blot analysis revealed decreases in the phosphorylation of heat shock protein 27 (HSP27), a downstream substrate of MK2. These results suggested that our peptides inhibited MK2 activity in vitro and should be investigated further as a potential therapeutic for applications involving inflammation. Furthermore, our results suggested that cell-penetrating peptides can be bioactive.
Collapse
Affiliation(s)
- Jamie L Brugnano
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Drive, West Lafayette, IN 47907, USA
| | | | | | | |
Collapse
|
34
|
A cell-penetrating peptide suppresses inflammation by inhibiting NF-κB signaling. Mol Ther 2011; 19:1849-57. [PMID: 21556052 DOI: 10.1038/mt.2011.82] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Nuclear factor-κB (NF-κB) is a central regulator of immune response and a potential target for developing anti-inflammatory agents. Mechanistic studies suggest that compounds that directly inhibit NF-κB DNA binding may block inflammation and the associated tissue damage. Thus, we attempted to discover peptides that could interfere with NF-κB signaling based on a highly conserved DNA-binding domain found in all NF-κB members. One such small peptide, designated as anti-inflammatory peptide-6 (AIP6), was characterized in the current study. AIP6 directly interacted with p65 and displayed an intrinsic cell-penetrating property. This peptide demonstrated significant anti-inflammatory effects in vitro and in vivo. In vitro, AIP6 inhibited the DNA-binding and transcriptional activities of the p65 NF-κB subunit as well as the production of inflammatory mediators in macrophages upon stimulation. Local administration of AIP6 significantly inhibited inflammation induced by zymosan in mice. Collectively, our results suggest that AIP6 is a promising lead peptide for the development of specific NF-κB inhibitors as potential anti-inflammatory agents.
Collapse
|
35
|
Ward BC, Kavalukas S, Brugnano J, Barbul A, Panitch A. Peptide inhibitors of MK2 show promise for inhibition of abdominal adhesions. J Surg Res 2011; 169:e27-36. [PMID: 21492875 DOI: 10.1016/j.jss.2011.01.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 11/24/2010] [Accepted: 01/21/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND Abdominal adhesions are a common side effect of surgical procedures with complications including infertility, chronic pain, and bowel obstruction, which may lead to the need for surgical lyses of the adhesions. Mitogen-activated protein kinase-activated protein kinase 2 (MK2) has been implicated in several diseases, involving inflammation and fibrosis. Thus, the development of a cell-penetrating peptide (CPP) that modulates MK2 activity may confer therapeutic benefit after abdominal surgery in general and more specifically after bowel anastomosis. METHODS This study evaluated the function of a CPP inhibitor of MK2 in human mesothelial cells and in a rat bowel anastomosis model. To determine IC50 and basic specificity, kinase inhibition was performed using a radiometric assay. Enzyme-linked immunoassay (ELISA) was used to evaluate interleukin-6 (IL-6) expression in response to IL-1β and tumor necrosis factor-α (TNF-α) stimulation in vitro to validate MK2 kinase inhibition. Following bowel anastomosis (10 rats for each control and treatment at 4 and 10 d), the rats were evaluated for weight loss, normal healing (colonic burst strength and hydroxyproline content at the anastomosis), and number and density of adhesions. RESULTS The IC50 of the MK2 inhibitor peptide (22 μM) was similar to that of the nonspecific small molecule rottlerin (IC50 = 5 μM). The MK2 inhibitor peptide was effective at suppressing IL-1β and TNF-α stimulated IL-6 expression in mesothelial cells. In vivo, the MK2 inhibitor peptide was effective at suppressing both the density and number of adhesions formed as a result of bowel an anastamosis. Importantly, the peptide had no negative effect on normal healing. CONCLUSIONS In conclusion, the peptide inhibitor of MK2, MMI-0100, has the potential to significantly reduce inflammation through suppression of inflammatory cytokine expression and showed promise as a therapeutic for abdominal adhesions.
Collapse
Affiliation(s)
- Brian C Ward
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907-2032, USA
| | | | | | | | | |
Collapse
|
36
|
Nain V, Sahi S, Verma A. CPP-ZFN: a potential DNA-targeting anti-malarial drug. Malar J 2010; 9:258. [PMID: 20846404 PMCID: PMC2949742 DOI: 10.1186/1475-2875-9-258] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 09/16/2010] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Multidrug-resistant Plasmodium is of major concern today. Effective vaccines or successful applications of RNAi-based strategies for the treatment of malaria are currently unavailable. An unexplored area in the field of malaria research is the development of DNA-targeting drugs that can specifically interact with parasitic DNA and introduce deleterious changes, leading to loss of vital genome function and parasite death. PRESENTATION OF THE HYPOTHESIS Advances in the development of zinc finger nuclease (ZFN) with engineered DNA recognition domains allow us to design and develop nuclease of high target sequence specificity with a mega recognition site that typically occurs only once in the genome. Moreover, cell-penetrating peptides (CPP) can cross the cell plasma membrane and deliver conjugated protein, nucleic acid, or any other cargo to the cytoplasm, nucleus, or mitochondria. This article proposes that a drug from the combination of the CPP and ZFN systems can effectively enter the intracellular parasite, introduce deleterious changes in its genome, and eliminate the parasite from the infected cells. TESTING THE HYPOTHESIS Availability of a DNA-binding motif for more than 45 triplets and its modular nature, with freedom to change number of fingers in a ZFN, makes development of customized ZFN against diverse target DNA sequence of any gene feasible. Since the Plasmodium genome is highly AT rich, there is considerable sequence site diversity even for the structurally and functionally conserved enzymes between Plasmodium and humans. CPP can be used to deliver ZFN to the intracellular nucleus of the parasite. Signal-peptide-based heterologous protein translocation to Plasmodium-infected RBCs (iRBCs) and different Plasmodium organelles have been achieved. With successful fusion of CPP with mitochondrial- and nuclear-targeting peptides, fusion of CPP with 1 more Plasmodium cell membrane translocation peptide seems achievable. IMPLICATIONS OF THE HYPOTHESIS Targeting of the Plasmodium genome using ZFN has great potential for the development of anti-malarial drugs. It allows the development of a single drug against all malarial infections, including multidrug-resistant strains. Availability of multiple ZFN target sites in a single gene will provide alternative drug target sites to combat the development of resistance in the future.
Collapse
Affiliation(s)
- Vikrant Nain
- School of Biotechnology, Gautam Buddha University, Greater Noida-201308, India
| | - Shakti Sahi
- School of Biotechnology, Gautam Buddha University, Greater Noida-201308, India
| | - Anju Verma
- School of Biological Sciences, University of Missouri, Kansas City, MO- 64110, USA
| |
Collapse
|
37
|
Bonduelle CV, Gillies ER. Dendritic Guanidines as Efficient Analogues of Cell Penetrating Peptides. Pharmaceuticals (Basel) 2010; 3:636-666. [PMID: 27713272 PMCID: PMC4033973 DOI: 10.3390/ph3030636] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Revised: 02/10/2010] [Accepted: 03/09/2010] [Indexed: 01/09/2023] Open
Abstract
The widespread application of cell penetrating agents to clinical therapeutics and imaging agents relies on the ability to prepare them on a large scale and to readily conjugate them to their cargos. Dendritic analogues of cell penetrating peptides, with multiple guanidine groups on their peripheries offer advantages as their high symmetry allows them to be efficiently synthesized, while orthogonal functionalities at their focal points allow them to be conjugated to cargo using simple synthetic methods. Their chemical structures and properties are also highly tunable as their flexibility and the number of guanidine groups can be tuned by altering the dendritic backbone or the linkages to the guanidine groups. This review describes the development of cell-penetrating dendrimers based on several different backbones, their structure-property relationships, and comparisons of their efficacies with those of known cell penetrating peptides. The toxicities of these dendritic guanidines are also reported as well as their application towards the intracellular delivery of biologically significant cargos including proteins and nanoparticles.
Collapse
Affiliation(s)
- Colin V Bonduelle
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, N6A 5B7, Canada
| | - Elizabeth R Gillies
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, N6A 5B7, Canada.
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, 1151 Richmond St., London, N6A 5B9, Canada.
| |
Collapse
|