1
|
Mirlohi K, Blocher McTigue WC. Coacervation for biomedical applications: innovations involving nucleic acids. SOFT MATTER 2024; 21:8-26. [PMID: 39641131 DOI: 10.1039/d4sm01253d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Gene therapies, drug delivery systems, vaccines, and many other therapeutics, although seeing breakthroughs over the past few decades, still suffer from poor stability, biocompatibility, and targeting. Coacervation, a liquid-liquid phase separation phenomenon, is a pivotal technique increasingly employed to enhance the effectiveness of therapeutics. Through coacervation strategies, many current challenges in therapeutic formulations can be addressed due to the tunable nature of this technique. However, much remains to be explored to enhance these strategies further and scale them from the benchtop to industrial applications. In this review, we highlight the underlying mechanisms of coacervation, elucidating how factors such as pH, ionic strength, temperature, chirality, and charge patterning influence the formation of coacervates and the encapsulation of active ingredients. We then present a perspective on current strategies harnessing these systems, specifically for nucleic acid-based therapeutics. These include peptide-, protein-, and polymer-based approaches, nanocarriers, and hybrid methods, each offering unique advantages and challenges. Nucleic acid-based therapeutics are crucial for designing rapid responses to diseases, particularly in pandemics. While these exciting systems offer many advantages, they also present limitations and challenges which are explored in this work. Exploring coacervation in the biomedical frontier opens new avenues for innovative nucleic acid-based treatments, marking a significant stride towards advanced therapeutic solutions.
Collapse
Affiliation(s)
- Kimiasadat Mirlohi
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA 18015, USA.
| | | |
Collapse
|
2
|
Omidi Y, Pourseif MM, Ansari RA, Barar J. Design and development of mRNA and self-amplifying mRNA vaccine nanoformulations. Nanomedicine (Lond) 2024; 19:2699-2725. [PMID: 39535127 DOI: 10.1080/17435889.2024.2419815] [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: 04/15/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
The rapid evolution of mRNA vaccines, highlighted by Pfizer-BioNTech and Moderna's COVID-19 vaccines, has transformed vaccine development and therapeutic approaches. Self-amplifying mRNA (saRNA) vaccines, a groundbreaking advancement in RNA-based vaccines, offer promising possibilities for disease prevention and treatment, including potential applications in cancer and neurodegenerative diseases. This review explores the complex design and development of these innovative vaccines, with a focus on their nanoscale formulations that utilize nanotechnology to improve their delivery and effectiveness. It articulates the fundamental principles of mRNA and saRNA vaccines, their mechanisms of action, and the role of synthetic mRNA in eliciting immune responses. The review further elaborates on various nanoscale delivery systems (e.g., lipid nanoparticles, polymeric nanoparticles and other nanocarriers), emphasizing their advantages in enhancing mRNA stability and cellular uptake. It addresses advanced nanoscale delivery techniques such as microfluidics and discusses the challenges in formulating mRNA and saRNA vaccines. By incorporating the latest technologies and current research, this review provides a thorough overview of recent mRNA and saRNA nanovaccines advancements, highlighting their potential to revolutionize vaccine technology and broaden clinical applications.
Collapse
Affiliation(s)
- Yadollah Omidi
- Department of Pharmaceutical Sciences, Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Mohammad M Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Engineered Biomaterial Research Center, Khazar University, Baku, Azerbaijan
| | - Rais A Ansari
- Department of Pharmaceutical Sciences, Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
3
|
Lim C, Blocher McTigue WC. Form Equals Function: Influence of Coacervate Architecture on Drug Delivery Applications. ACS Biomater Sci Eng 2024; 10:6766-6789. [PMID: 39423330 PMCID: PMC11558567 DOI: 10.1021/acsbiomaterials.4c01105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024]
Abstract
Complex coacervates, formed through electrostatic interactions between oppositely charged polymers, present a versatile platform for drug delivery, providing rapid assembly, selective encapsulation, and responsiveness to environmental stimuli. The architecture and properties of coacervates can be tuned by controlling structural and environmental design factors, which significantly impact the stability and delivery efficiency of the drugs. While environmental design factors such as salt, pH, and temperature play a crucial role in coacervate formation, structural design factors such as polymer concentration, polymer structure, mixing ratio, and chain length serve as the core framework that shapes coacervate architecture. These elements modulate the phase behavior and material properties of coacervates, allowing for a highly tunable system. In this review, we primarily analyze how these structural design factors contribute to the formation of diverse coacervate architecture, ranging from bulk coacervates to polyion complex micelles, vesicles, and cross-linked gels, though environmental design factors are considered. We then examine the effectiveness of these architectures in enhancing the delivery and efficacy of drugs across various administration routes, such as noninvasive (e.g., oral and transdermal) and invasive delivery. This review aims to provide foundational insights into the design of advanced drug delivery systems by examining how the origin and chemical structure of polymers influence coacervate architecture, which in turn defines their material properties. We then explore how the architecture can be tailored to optimize drug delivery for specific administration routes. This approach leverages the intrinsic properties derived from the coacervate architecture to enable targeted, controlled, and efficient drug release, ultimately enhancing therapeutic outcomes in precision medicine.
Collapse
Affiliation(s)
- Chaeyoung Lim
- Department of Chemical and Biomolecular
Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Whitney C. Blocher McTigue
- Department of Chemical and Biomolecular
Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
4
|
Zhang R, Rygelski BT, Kruse LE, Smith JD, Wang X, Allen BN, Kramer JS, Seim GF, Faulkner TJ, Kuang H, Kokkoli E, Schrum AG, Ulery BD. Adjuvant Delivery Method and Nanoparticle Charge Influence Peptide Amphiphile Micelle Vaccine Bioactivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598369. [PMID: 38915689 PMCID: PMC11195052 DOI: 10.1101/2024.06.10.598369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Vaccines are an indispensable public health measure that have enabled the eradication, near elimination, and prevention of a variety of pathogens. As research continues and our understanding of immunization strategies develops, subunit vaccines have emerged as exciting alternatives to existing whole vaccine approaches. Unfortunately, subunit vaccines often possess weak antigenicity, requiring delivery devices and adjuvant supplementation to improve their utility. Peptide amphiphile micelles have recently been shown to function as both delivery devices and self-adjuvanting systems that can be readily associated with molecular adjuvants to further improve vaccine-mediated host immunity. While promising, many design rules associated with the plethora of underlying adjustable parameters in the generation of a peptide amphiphile micelle vaccine have yet to be uncovered. This work explores the impact micellar adjuvant complexation method and incorporated antigen type have on their ability to activate dendritic cells and induce antigen specific responses. Interestingly, electrostatic complexation of CpG to micelles resulted in improved in vitro dendritic cell activation over hydrophobic association and antigen|adjuvant co-localization influenced cell-mediated, but not antibody-mediated immune responses. These exciting results complement those previously published to build the framework of a micelle vaccine toolbox that can be leveraged for future disease specific formulations.
Collapse
|
5
|
Huynh TP, Wittig NKL, Andersen A, Bach-Gansmo FL, Birkedal H. Underwater Fabrication of Carbon Nanotube/Coacervate Composites. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:13010-13016. [PMID: 38858173 DOI: 10.1021/acs.langmuir.4c00715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Soft conductive materials are of interest for a wide range of applications, but their syntheses have remained difficult. Herein, we present a convenient route for underwater fabrication of a composite made of carbon nanotubes (CNTs) and mussel-inspired complex coacervates. The key to success of this method is that CNTs were solubilized very effectively in protocoacervates, which are high-concentration solutions of polyelectrolytes at a pH where only one of them is charged, thereby impeding coacervate formation. Composite materials were formed by the simple injection of CNT-dispersed protocoacervate solutions into water under ambient conditions. The method is simple, fast, and ecofriendly, and composites of CNT-complex coacervate in the form of films or bulk materials were obtained. The composites demonstrated electrical conductivity and tunable mechanical properties, which depended on the concentration of polyelectrolytes and the CNT:protocoacervate ratio. Hence, the composites can be manipulated to attain diverse properties, for examples, tunable reduced modulus (15 to 32 GPa) and hardness (0.3 to 0.7 GPa) as well as an electrical conductivity of up to 4 × 103 S m-1.
Collapse
Affiliation(s)
- Tan-Phat Huynh
- Department of Chemistry & iNANO, Aarhus University, 14 Gustav Wieds Vej, Aarhus 8000, Denmark
| | - Nina Ko Lln Wittig
- Department of Chemistry & iNANO, Aarhus University, 14 Gustav Wieds Vej, Aarhus 8000, Denmark
| | - Amanda Andersen
- Department of Chemistry & iNANO, Aarhus University, 14 Gustav Wieds Vej, Aarhus 8000, Denmark
| | | | - Henrik Birkedal
- Department of Chemistry & iNANO, Aarhus University, 14 Gustav Wieds Vej, Aarhus 8000, Denmark
| |
Collapse
|
6
|
Roy PS. Complex Coacervate-Based Materials for Biomedicine: Recent Advancements and Future Prospects. Ind Eng Chem Res 2024; 63:5414-5487. [DOI: 10.1021/acs.iecr.3c03830] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Partha Sarathi Roy
- Division of Pharmaceutical Sciences, Health Sciences Building, University of Missouri─Kansas City, 2464 Charlotte St., Kansas City, Missouri 64108-2718, United States
- Department of Pharmaceutics/Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, 751 Brookside Rd., Stockton, California 95211, United States
| |
Collapse
|
7
|
Forenzo C, Larsen J. Complex Coacervates as a Promising Vehicle for mRNA Delivery: A Comprehensive Review of Recent Advances and Challenges. Mol Pharm 2023; 20:4387-4403. [PMID: 37561647 DOI: 10.1021/acs.molpharmaceut.3c00439] [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] [Indexed: 08/12/2023]
Abstract
Messenger RNA (mRNA)-based therapies have gained significant attention, following the successful deployment of mRNA-based COVID-19 vaccines. Compared with traditional methods of genetic modification, mRNA-based therapies offer several advantages, including a lower risk of genetic mutations, temporary and controlled therapeutic gene expression, and a shorter production time, which facilitates rapid responses to emerging health challenges. Moreover, mRNA-based therapies have shown immense potential in treating a wide range of diseases including cancers, immune diseases, and neurological disorders. However, the current limitations of non-viral vectors for efficient and safe delivery of mRNA therapies, such as low encapsulation efficiency, potential toxicity, and limited stability, necessitate the exploration of novel strategies to overcome these challenges and fully realize the potential of mRNA-based therapeutics. Coacervate-based delivery systems have recently emerged as promising strategies for enhancing mRNA delivery. Coacervates, which are formed by the aggregation of two or more macromolecules, have shown great potential in delivering a wide range of therapeutics due to their ability to form a separated macromolecular-rich fluid phase in an aqueous environment. This phase separation enables the entrapment and protection of therapeutic agents from degradation as well as efficient cellular uptake and controlled release. Additionally, the natural affinity of coacervates for mRNA molecules presents an excellent opportunity for enhancing mRNA delivery to targeted cells and tissues, making coacervate-based delivery systems an attractive option for mRNA-based therapies. This review highlights the limitations of current strategies for mRNA delivery and the advantages of coacervate-based delivery systems to enable mRNA therapeutics. Coacervates protect mRNA from enzymatic degradation and enhance cellular uptake, leading to sustained and controlled gene expression. Despite their promising properties, the specific use of coacervates as mRNA delivery vehicles remains underexplored. This review aims to provide a comprehensive overview of coacervate-mediated delivery of mRNA, exploring the properties and applications of different coacervating agents as well as the challenges and optimization strategies involved in mRNA encapsulation, release, stability, and translation via coacervate-mediated delivery. Through a comprehensive analysis of recent advancements and recommended future directions, our review sheds light on the promising role of coacervate-mediated delivery for RNA therapeutics, highlighting its potential to enable groundbreaking applications in drug delivery and gene therapy.
Collapse
|
8
|
Choi J, Heo T, Choi H, Choi S, Won J. Co‐assembly
behavior of oppositely charged thermoresponsive elastin‐like polypeptide block copolymers. J Appl Polym Sci 2022. [DOI: 10.1002/app.52906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jeong‐Wan Choi
- Department of Chemical Engineering Hongik University Seoul Republic of Korea
| | - Tae‐Young Heo
- Department of Chemical Engineering Hongik University Seoul Republic of Korea
| | - Heelak Choi
- Department of Chemical Engineering Hongik University Seoul Republic of Korea
| | - Soo‐Hyung Choi
- Department of Chemical Engineering Hongik University Seoul Republic of Korea
| | - Jong‐In Won
- Department of Chemical Engineering Hongik University Seoul Republic of Korea
| |
Collapse
|
9
|
Ban E, Kim A. Coacervates: recent developments as nanostructure delivery platforms for therapeutic biomolecules. Int J Pharm 2022; 624:122058. [PMID: 35905931 DOI: 10.1016/j.ijpharm.2022.122058] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 10/16/2022]
Abstract
Coacervation is a liquid-liquid phase separation that can occur in solutions of macromolecules through self-assembly or electrostatic interactions. Recently, coacervates composed of biocompatible macromolecules have been actively investigated as nanostructure platforms to encapsulate and deliver biomolecules such as proteins, RNAs, and DNAs. One particular advantage of coacervates is that they are derived from aqueous solutions, unlike other nanoparticle delivery systems that often require organic solvents. In addition, coacervates achieve high loading while maintaining the viability of the cargo material. Here, we review recent developments in the applications of coacervates and their limitations in the delivery of therapeutic biomolecules. Important factors for coacervation include molecular structures of the polyelectrolytes, mixing ratio, the concentration of polyelectrolytes, and reaction conditions such as ionic strength, pH, and temperature. Various compositions of coacervates have been shown to deliver biomolecules in vitro and in vivo with encouraging activities. However, major hurdles remain for the systemic route of administration other than topical or local delivery. The scale-up of manufacturing methods suitable for preclinical and clinical evaluations remains to be addressed. We conclude with a few research directions to overcome current challenges, which may lead to successful translation into the clinic.
Collapse
Affiliation(s)
- Eunmi Ban
- College of Pharmacy, CHA University, Seongnam 13488, Korea
| | - Aeri Kim
- College of Pharmacy, CHA University, Seongnam 13488, Korea.
| |
Collapse
|
10
|
Humphries B, Wang Z, Yang C. MicroRNA Regulation of Breast Cancer Stemness. Int J Mol Sci 2021; 22:3756. [PMID: 33916548 PMCID: PMC8038508 DOI: 10.3390/ijms22073756] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 12/22/2022] Open
Abstract
Recent advances in our understanding of breast cancer have demonstrated that cancer stem-like cells (CSCs, also known as tumor-initiating cell (TICs)) are central for progression and recurrence. CSCs are a small subpopulation of cells present in breast tumors that contribute to growth, metastasis, therapy resistance, and recurrence, leading to poor clinical outcome. Data have shown that cancer cells can gain characteristics of CSCs, or stemness, through alterations in key signaling pathways. The dysregulation of miRNA expression and signaling have been well-documented in cancer, and recent studies have shown that miRNAs are associated with breast cancer initiation, progression, and recurrence through regulating CSC characteristics. More specifically, miRNAs directly target central signaling nodes within pathways that can drive the formation, maintenance, and even inhibition of the CSC population. This review aims to summarize these research findings specifically in the context of breast cancer. This review also discusses miRNAs as biomarkers and promising clinical therapeutics, and presents a comprehensive summary of currently validated targets involved in CSC-specific signaling pathways in breast cancer.
Collapse
Affiliation(s)
- Brock Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Zhishan Wang
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Drive, Cleveland, OH 44109, USA;
| | - Chengfeng Yang
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Drive, Cleveland, OH 44109, USA;
| |
Collapse
|
11
|
Mintis DG, Mavrantzas VG. Phase Boundary and Salt Partitioning in Coacervate Complexes Formed between Poly(acrylic acid) and Poly(N,N-dimethylaminoethyl methacrylate) from Detailed Atomistic Simulations Combined with Free Energy Perturbation and Thermodynamic Integration Calculations. Macromolecules 2020. [DOI: 10.1021/acs.macromol.0c00728] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Dimitris G. Mintis
- Department of Chemical Engineering, University of Patras & FORTH-ICE/HT, Patras GR26504, Greece
| | - Vlasis G. Mavrantzas
- Department of Chemical Engineering, University of Patras & FORTH-ICE/HT, Patras GR26504, Greece
- Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zürich, CH-8092 Zürich, Switzerland
| |
Collapse
|
12
|
Humphries BA, Wang Z, Yang C. MicroRNA Regulation of the Small Rho GTPase Regulators-Complexities and Opportunities in Targeting Cancer Metastasis. Cancers (Basel) 2020; 12:E1092. [PMID: 32353968 PMCID: PMC7281527 DOI: 10.3390/cancers12051092] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 02/07/2023] Open
Abstract
The small Rho GTPases regulate important cellular processes that affect cancer metastasis, such as cell survival and proliferation, actin dynamics, adhesion, migration, invasion and transcriptional activation. The Rho GTPases function as molecular switches cycling between an active GTP-bound and inactive guanosine diphosphate (GDP)-bound conformation. It is known that Rho GTPase activities are mainly regulated by guanine nucleotide exchange factors (RhoGEFs), GTPase-activating proteins (RhoGAPs), GDP dissociation inhibitors (RhoGDIs) and guanine nucleotide exchange modifiers (GEMs). These Rho GTPase regulators are often dysregulated in cancer; however, the underlying mechanisms are not well understood. MicroRNAs (miRNAs), a large family of small non-coding RNAs that negatively regulate protein-coding gene expression, have been shown to play important roles in cancer metastasis. Recent studies showed that miRNAs are capable of directly targeting RhoGAPs, RhoGEFs, and RhoGDIs, and regulate the activities of Rho GTPases. This not only provides new evidence for the critical role of miRNA dysregulation in cancer metastasis, it also reveals novel mechanisms for Rho GTPase regulation. This review summarizes recent exciting findings showing that miRNAs play important roles in regulating Rho GTPase regulators (RhoGEFs, RhoGAPs, RhoGDIs), thus affecting Rho GTPase activities and cancer metastasis. The potential opportunities and challenges for targeting miRNAs and Rho GTPase regulators in treating cancer metastasis are also discussed. A comprehensive list of the currently validated miRNA-targeting of small Rho GTPase regulators is presented as a reference resource.
Collapse
Affiliation(s)
- Brock A. Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Zhishan Wang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 V A Drive, Lexington, KY 40536, USA;
| | - Chengfeng Yang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 V A Drive, Lexington, KY 40536, USA;
| |
Collapse
|
13
|
Fluorescent complex coacervates of agar and in situ formed zein nanoparticles: Role of electrostatic forces. Carbohydr Polym 2019; 224:115150. [DOI: 10.1016/j.carbpol.2019.115150] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 12/16/2022]
|
14
|
Humphries B, Wang Z, Yang C. MicroRNA Regulation of Epigenetic Modifiers in Breast Cancer. Cancers (Basel) 2019; 11:E897. [PMID: 31252590 PMCID: PMC6678197 DOI: 10.3390/cancers11070897] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/16/2019] [Accepted: 06/24/2019] [Indexed: 12/20/2022] Open
Abstract
Epigenetics refers to the heritable changes in gene expression without a change in the DNA sequence itself. Two of these major changes include aberrant DNA methylation as well as changes to histone modification patterns. Alterations to the epigenome can drive expression of oncogenes and suppression of tumor suppressors, resulting in tumorigenesis and cancer progression. In addition to modifications of the epigenome, microRNA (miRNA) dysregulation is also a hallmark for cancer initiation and metastasis. Advances in our understanding of cancer biology demonstrate that alterations in the epigenome are not only a major cause of miRNA dysregulation in cancer, but that miRNAs themselves also indirectly drive these DNA and histone modifications. More explicitly, recent work has shown that miRNAs can regulate chromatin structure and gene expression by directly targeting key enzymes involved in these processes. This review aims to summarize these research findings specifically in the context of breast cancer. This review also discusses miRNAs as epigenetic biomarkers and as therapeutics, and presents a comprehensive summary of currently validated epigenetic targets in breast cancer.
Collapse
Affiliation(s)
- Brock Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI 48109; USA.
| | - Zhishan Wang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Chengfeng Yang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA.
- Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY 40536; USA.
| |
Collapse
|
15
|
Zhang Y, Ma J, Xu Q. Polyelectrolyte complex from cationized casein and sodium alginate for fragrance controlled release. Colloids Surf B Biointerfaces 2019; 178:439-444. [DOI: 10.1016/j.colsurfb.2019.03.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 03/07/2019] [Indexed: 11/16/2022]
|
16
|
Wang Z, Li Y, Xiao Y, Lin HP, Yang P, Humphries B, Gao T, Yang C. Integrin α9 depletion promotes β-catenin degradation to suppress triple-negative breast cancer tumor growth and metastasis. Int J Cancer 2019; 145:2767-2780. [PMID: 31008533 DOI: 10.1002/ijc.32359] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/26/2019] [Accepted: 04/16/2019] [Indexed: 12/27/2022]
Abstract
Although integrin α9 (ITGA9) is known to be involved in cell adhesion and motility, its expression in cancer and its role in tumor growth and metastasis remain largely unknown. Our study was designed to investigate the role of ITGA9 in triple-negative breast cancer (TNBC). ITGA9 expression in TNBC cells was knocked out (KO) using CRISPR/Cas9 technology. Four orthotopic mouse mammary xenograft tumor models coupled with cell culture studies were performed to determine the effect of ITGA9 depletion on TNBC tumor growth and metastasis and the underlying mechanism. Bioinformatics analysis showed that ITGA9 level is significantly higher in TNBC than other breast cancer subtypes, and higher ITGA9 level is associated with significantly worse distant metastasis-free survival and recurrence-free survival in TNBC patients. Experimentally, ITGA9 KO significantly reduced TNBC cell cancer stem cell (CSC)-like property, tumor angiogenesis, tumor growth and metastasis by promoting β-catenin degradation. Further mechanistic studies revealed that ITGA9 KO causes integrin-linked kinase (ILK) relocation from the membrane region to the cytoplasm, where it interacts with protein kinase A (PKA) and inhibits PKA activity leading to increased activity of glycogen synthase kinase 3 (GSK3) and subsequent β-catenin degradation. Overexpressing β-catenin in ITGA9 KO cells reversed the inhibitory effect of ITGA9 KO on tumor growth and metastasis. Furthermore, ITGA9 downregulation in TNBC tumors by nanoparticle-mediated delivery of ITGA9 siRNA drastically decreased tumor angiogenesis, tumor growth and metastasis. These findings indicate that ITGA9 depletion suppresses TNBC tumor growth and metastasis by promoting β-catenin degradation through the ILK/PKA/GSK3 pathway.
Collapse
Affiliation(s)
- Zhishan Wang
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY
| | - Yunfei Li
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY
| | - Yajuan Xiao
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY.,Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Hsuan-Pei Lin
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY
| | - Ping Yang
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY.,School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Brock Humphries
- Department of Radiology, University of Michigan, Ann Arbor, MI
| | - Tianyan Gao
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY
| | - Chengfeng Yang
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY
| |
Collapse
|
17
|
Beals N, Kasibhatla N, Basu S. Efficient Delivery of Plasmid DNA Using Incorporated Nucleotides for Precise Conjugation of Targeted Nanoparticles. ACS APPLIED BIO MATERIALS 2019; 2:717-727. [DOI: 10.1021/acsabm.8b00596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Nathan Beals
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44242, United States
| | - Nithya Kasibhatla
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44242, United States
| | - Soumitra Basu
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44242, United States
| |
Collapse
|
18
|
Liang Q, Li F, Li Y, Liu Y, Lan M, Wu S, Wu X, Ji Y, Zhang R, Yin L. Self-assisted membrane-penetrating helical polypeptides mediate anti-inflammatory RNAi against myocardial ischemic reperfusion (IR) injury. Biomater Sci 2019; 7:3717-3728. [DOI: 10.1039/c9bm00719a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Aromatically-modified helical polypeptide mediates membrane-penetrating RAGE siRNA delivery toward anti-inflammatory treatment against myocardial IR injury.
Collapse
|
19
|
Huang L, Chen J, He M, Hou X, Lu Y, Lou K, Gao F. Nanoparticle structure transformation of mPEG grafted chitosan with rigid backbone induced by α-cyclodextrin. CHINESE CHEM LETT 2019. [DOI: 10.1016/j.cclet.2017.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
20
|
Vecchies F, Sacco P, Decleva E, Menegazzi R, Porrelli D, Donati I, Turco G, Paoletti S, Marsich E. Complex Coacervates between a Lactose-Modified Chitosan and Hyaluronic Acid as Radical-Scavenging Drug Carriers. Biomacromolecules 2018; 19:3936-3944. [PMID: 30204431 DOI: 10.1021/acs.biomac.8b00863] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Complex coacervation of two oppositely charged polysaccharides, namely a lactose-modified chitosan (CTL) and hyaluronan (HA), was investigated in this study. Coacervates of the two polysaccharides were prepared by drop-by-drop injection of HA into CTL. Transmittance and dynamic light scattering (DLS) measurements in combination with TEM analyses demonstrated the formation of spheroidal colloids in the nano-/microsize range showing good homogeneity. Strikingly, the presence of 150 mM supporting NaCl did not hamper the colloid formation. Stability studies on selected formulations demonstrated that HA/CTL coacervates were stable up to 3 weeks at 37 °C and behaved as pH-responsive colloids since transition from entangled to disentangled chains was attained for a proper pH range. The possibility of freeze-drying the coacervates for storage purposes and the ability of encapsulating selected payloads were investigated as well, for two values of the fraction of the lactitol side-chain substitution (FL). Finally, biological tests using human neutrophils were undertaken at acidic pH value (pH = 6.0): under such experimental conditions, akin to those frequently occurring in the inflammatory microenvironment, coacervates scavenged reactive oxygen species (ROS) generated by these cells in basal conditions. Given the well documented bioactivity of CTL with respect to chitosan toward cartilage regeneration, these findings point to a possible application of HA/CTL-based colloids as scavenging and bioactive carriers for the delivery of therapeutic molecules at confined inflamed sites such as knee joints.
Collapse
Affiliation(s)
- Federica Vecchies
- Department of Life Sciences , University of Trieste , Via L. Giorgieri 5 , I-34127 Trieste , Italy
| | - Pasquale Sacco
- Department of Life Sciences , University of Trieste , Via L. Giorgieri 5 , I-34127 Trieste , Italy
| | - Eva Decleva
- Department of Life Sciences , University of Trieste , Via L. Giorgieri 5 , I-34127 Trieste , Italy
| | - Renzo Menegazzi
- Department of Life Sciences , University of Trieste , Via L. Giorgieri 5 , I-34127 Trieste , Italy
| | - Davide Porrelli
- Department of Medicine, Surgery and Health Sciences , University of Trieste , Piazza dell'Ospitale 1 , I-34125 Trieste , Italy
| | - Ivan Donati
- Department of Life Sciences , University of Trieste , Via L. Giorgieri 5 , I-34127 Trieste , Italy
| | - Gianluca Turco
- Department of Medicine, Surgery and Health Sciences , University of Trieste , Piazza dell'Ospitale 1 , I-34125 Trieste , Italy
| | - Sergio Paoletti
- Department of Life Sciences , University of Trieste , Via L. Giorgieri 5 , I-34127 Trieste , Italy
| | - Eleonora Marsich
- Department of Medicine, Surgery and Health Sciences , University of Trieste , Piazza dell'Ospitale 1 , I-34125 Trieste , Italy
| |
Collapse
|
21
|
Benner NL, Near KE, Bachmann MH, Contag CH, Waymouth RM, Wender PA. Functional DNA Delivery Enabled by Lipid-Modified Charge-Altering Releasable Transporters (CARTs). Biomacromolecules 2018; 19:2812-2824. [PMID: 29727572 PMCID: PMC6542359 DOI: 10.1021/acs.biomac.8b00401] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Safe and effective DNA delivery systems are required to enable or enhance clinical strategies and research involving gene therapy and DNA vaccinations. To address this delivery problem, a series of charge-altering releasable transporters (CARTs) with varied lipid content were prepared and evaluated for plasmid DNA (pDNA) delivery into cultured cells. These lipid-modified CART co-oligomers were synthesized in only two steps via sequential organocatalytic ring-opening polymerization of lipid-containing cyclic carbonate monomers and morpholinone monomers. Lipid variations of the CARTs substantially impacted the delivery efficiency of pDNA, with oleyl- and linoleyl-based CARTs showing enhanced performance relative to the commercial transfection agent Lipofectamine 2000 (L2000). The best-performing oleyl CART was carried forward to study stable luciferase transfection with a Sleeping Beauty ( SB) transposon system. The oleyl CART outperformed the L2000 positive control with respect to stable transfection efficiency. CART-pDNA complexes represent a new DNA delivery system for research and clinical applications.
Collapse
Affiliation(s)
- Nancy L. Benner
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Katherine E. Near
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Michael H. Bachmann
- Department of Pediatrics, Stanford University, Stanford, California 94305, United States
| | - Christopher H. Contag
- Department of Pediatrics, Stanford University, Stanford, California 94305, United States
- Department of Microbiology and Immunology, Stanford University, Stanford, California 94305, United States
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Robert M. Waymouth
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Department of Chemical and Systems Biology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
22
|
Li Y, Humphries B, Yang C, Wang Z. Nanoparticle-Mediated Therapeutic Agent Delivery for Treating Metastatic Breast Cancer-Challenges and Opportunities. NANOMATERIALS 2018; 8:nano8060361. [PMID: 29794968 PMCID: PMC6027372 DOI: 10.3390/nano8060361] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 05/21/2018] [Accepted: 05/22/2018] [Indexed: 12/19/2022]
Abstract
Breast cancer (BC) is the second leading cause of cancer-related death in American women and more than 90% of BC-related death is caused by metastatic BC (MBC). This review stresses the limited success of traditional therapies as well as the use of nanomedicine for treating MBC. Understanding the biological barriers of MBC that nanoparticle in vivo trafficking must overcome could provide valuable new insights for translating nanomedicine from the bench side to the bedside. A view about nanomedicine applied in BC therapy has been summarized with their present status, which is gaining attention in the clinically-applied landscape. The progressions of drug/gene delivery systems, especially the status of their preclinical or clinical trials, are also discussed. Here we highlight that the treatment of metastasis, in addition to the extensively described inhibition of primary tumor growth, is an indispensable requirement for nanomedicine. Along with more innovations in material chemistry and more progressions in biology, nanomedicine will constantly supply more exciting new approaches for targeted drug/gene delivery against MBC.
Collapse
Affiliation(s)
- Yunfei Li
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA.
- Department of Pharmaceutics, Institute of Medicinal Biotechnology, Peking Union Medical College, Beijing 100050, China.
| | - Brock Humphries
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Chengfeng Yang
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA.
| | - Zhishan Wang
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
23
|
Xu X, Li Y, Liang Q, Song Z, Li F, He H, Wang J, Zhu L, Lin Z, Yin L. Efficient Gene Delivery Mediated by a Helical Polypeptide: Controlling the Membrane Activity via Multivalency and Light-Assisted Photochemical Internalization (PCI). ACS APPLIED MATERIALS & INTERFACES 2018; 10:256-266. [PMID: 29206023 DOI: 10.1021/acsami.7b15896] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The development of robust and nontoxic membrane-penetrating materials is highly demanded for nonviral gene delivery. Herein, a photosensitizer (PS)-embedded, star-shaped helical polypeptide was developed, which combines the advantages of multivalency-enhanced intracellular DNA uptake and light-strengthened endosomal escape to enable highly efficient gene delivery with low toxicity. 5,10,15,20-Tetrakis-(4-aminophenyl) porphyrin as a selected PS initiated ring-opening polymerization of N-carboxyanhydride and yielded a star-shaped helical polypeptide after side-chain functionalization with guanidine groups. The star polypeptide afforded a notably higher transfection efficiency and lower cytotoxicity than those of its linear analogue. Light irradiation caused almost complete (∼90%) endosomal release of the DNA cargo via the photochemical internalization (PCI) mechanism and further led to a 6-8-fold increment of the transfection efficiency in HeLa, B16F10, and RAW 264.7 cells, outperforming commercial reagent 25k PEI by up to 3 orders of magnitude. Because the PS and DNA cargoes were compartmentalized distantly in the core and polypeptide layers, respectively, the generated reactive oxygen species caused minimal damage to DNA molecules to preserve their transfection potency. Such multivalency- and PCI-potentiated gene delivery efficiency was also demonstrated in vivo in melanoma-bearing mice. This study thus provides a promising strategy to overcome the multiple membrane barriers against nonviral gene delivery.
Collapse
Affiliation(s)
- Xin Xu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University , Suzhou 215123, China
| | - Yongjuan Li
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University , Suzhou 215123, China
| | - Qiujun Liang
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University , Suzhou 215123, China
| | - Ziyuan Song
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign , 1304 W Green Street, Urbana, Illinois 61801, United States
| | - Fangfang Li
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University , Suzhou 215123, China
| | - Hua He
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University , Suzhou 215123, China
| | - Jinhui Wang
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University , Suzhou 215123, China
| | - Lipeng Zhu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University , Suzhou 215123, China
| | - Zhifeng Lin
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University of Medicine , Shanghai 200080, China
| | - Lichen Yin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University , Suzhou 215123, China
| |
Collapse
|
24
|
Zhu L, Simpson JM, Xu X, He H, Zhang D, Yin L. Cationic Polypeptoids with Optimized Molecular Characteristics toward Efficient Nonviral Gene Delivery. ACS APPLIED MATERIALS & INTERFACES 2017; 9:23476-23486. [PMID: 28653538 DOI: 10.1021/acsami.7b06031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The rational design of gene vectors relies on the understanding of their structure-property relationship. Polypeptoids, which are structural isomers of natural polypeptides, hold great potential as gene delivery vectors due to their facile preparation, structural tunability, and most importantly, their desirable proteolytic stability. We herein designed a library of polypeptoids with different cationic side-chain terminal groups, degree of polymerizations (DPs), side-chain lengths, and incorporated aliphatic side chains, to unravel the structure-property relationships so that gene delivery efficiency can be maximized and cytotoxicity can be minimized. In HeLa cells, a polypeptoid bearing a primary amine side-chain terminal group exhibited remarkably higher transfection efficiency than that of its analogues containing secondary, tertiary, or quaternary amine groups. Elongation of the polypeptoid backbone length (from 28 to 251 mer) led to enhanced DNA condensation as well as cellular uptake levels, however it also caused higher cytotoxicity. Upon a proper balance between DNA uptake and cytotoxicity, the polypeptoid with a DP of 46 afforded the highest transfection efficiency. Elongating the aliphatic spacer between the backbone and side amine groups enhanced the hydrophobicity of the side chains, which resulted in notably increased membrane activities and transfection efficiency. Further incorporation of hydrophobic decyl side chains led to an improvement in transfection efficiency of ∼6 fold. The top-performing material identified, P11, mediated successful gene transfection under serum-containing conditions, outperforming the commercial transfection reagent poly(ethylenimine) by nearly 4 orders of magnitude. Reflecting its excellent serum-resistant properties, P11 further enabled effective transfection in vivo following intratumoral injection to melanoma-bearing mice. This study will help the rational design of polypeptoid-based gene delivery materials, and the best-performing material identified may provide a potential supplement to existing gene vectors.
Collapse
Affiliation(s)
- Lipeng Zhu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University , Suzhou 215123, P. R. China
| | - Jessica M Simpson
- Department of Chemistry and Macromolecular Studies Group, Louisiana State University , Baton Rouge, Louisiana 70803, United States
| | - Xin Xu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University , Suzhou 215123, P. R. China
| | - Hua He
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University , Suzhou 215123, P. R. China
| | - Donghui Zhang
- Department of Chemistry and Macromolecular Studies Group, Louisiana State University , Baton Rouge, Louisiana 70803, United States
| | - Lichen Yin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University , Suzhou 215123, P. R. China
| |
Collapse
|