Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(24): eade9488, 2023 06 16.
Article in English | MEDLINE | ID: mdl-37327334

ABSTRACT

Biomedical devices comprise a major component of modern medicine, however immune-mediated fibrosis and rejection can limit their function over time. Here, we describe a humanized mouse model that recapitulates fibrosis following biomaterial implantation. Cellular and cytokine responses to multiple biomaterials were evaluated across different implant sites. Human innate immune macrophages were verified as essential to biomaterial rejection in this model and were capable of cross-talk with mouse fibroblasts for collagen matrix deposition. Cytokine and cytokine receptor array analysis confirmed core signaling in the fibrotic cascade. Foreign body giant cell formation, often unobserved in mice, was also prominent. Last, high-resolution microscopy coupled with multiplexed antibody capture digital profiling analysis supplied spatial resolution of rejection responses. This model enables the study of human immune cell-mediated fibrosis and interactions with implanted biomaterials and devices.


Subject(s)
Biocompatible Materials , Foreign Bodies , Humans , Animals , Mice , Foreign-Body Reaction/etiology , Disease Models, Animal , Cytokines , Fibrosis
2.
Bioconjug Chem ; 33(7): 1279-1285, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35758018

ABSTRACT

The indiscriminate biodistribution of therapeutics can be a key barrier to their safety and efficacy. Localization of compounds into non-diseased tissues often leads to both toxic and dose-limiting effects. To overcome this barrier, nanomedicine implements targeting agents to localize or selectively uptake drugs at disease sites. However, to date there are only a small number of targeting agents with limited scope for targeting tissues. Small-molecule ligands are particularly attractive as targeting agents due to their relatively low cost, tunability, and ease of conjugation. Currently, there are no systematic approaches to the discovery of new small-molecule targeting ligands. Here, we developed a quantitative metal-encoded conjugate platform to determine the biodistribution of multiple small molecules in vivo. By utilizing lanthanide metal complexes, this platform successfully distinguished known ligands with differential tissue targeting in vivo. This system will facilitate the discovery of small molecules as targeting ligands and can accelerate the identification of novel biological targets for tissue-targeted drug delivery.


Subject(s)
Drug Delivery Systems , Nanomedicine , Ligands , Pharmaceutical Preparations , Tissue Distribution
3.
Nano Lett ; 21(18): 7495-7504, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34495662

ABSTRACT

Structured nanoassemblies are biomimetic structures that are enabling applications from nanomedicine to catalysis. One approach to achieve these spatially organized architectures is utilizing amphiphilic diblock copolymers with one or two macromolecular backbones that self-assemble in solution. To date, the impact of alternating backbone architectures on self-assembly and drug delivery is still an area of active research limited by the strategies used to synthesize these multiblock polymers. Here, we report self-assembling ABC-type alginate-based triblock copolymers with the backbones of three distinct biomaterials utilizing a facile conjugation approach. This "polymer mosaic" was synthesized by the covalent attachment of alginate with a PLA/PEG diblock copolymer. The combination of alginate, PEG, and PLA domains resulted in an amphiphilic copolymer that self-assembles into nanoparticles with a unique morphology of alginate domain compartmentalization. These particles serve as a versatile platform for co-encapsulation of hydrophilic and hydrophobic small molecules, their spatiotemporal release, and show potential as a drug delivery system for combination therapy.


Subject(s)
Alginates , Micelles , Hydrophobic and Hydrophilic Interactions , Polyethylene Glycols , Polymers
5.
ACS Chem Biol ; 15(10): 2649-2654, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32902255

ABSTRACT

Interleukin-4 (IL-4) is a multifunctional cytokine and an important regulator of inflammation. When deregulated, IL-4 activity is associated with asthma, allergic inflammation, and multiple types of cancer. While antibody-based inhibitors targeting the soluble cytokine have been evaluated clinically, they failed to achieve their end points in trials. Small-molecule inhibitors are an attractive alternative, but identifying effective chemotypes that inhibit the protein-protein interactions between cytokines and their receptors remains an active area of research. As a result, no small-molecule inhibitors to the soluble IL-4 cytokine have yet been reported. Here, we describe the first IL-4 small-molecule inhibitor identified and characterized through a combination of binding-based approaches and cell-based activity assays. The compound features a nicotinonitrile scaffold with micromolar affinity and potency for the cytokine and disrupts type II IL-4 signaling in cells. Small-molecule inhibitors of these important cell-signaling proteins have implications for numerous immune-related disorders and inform future drug discovery and design efforts for these challenging protein targets.


Subject(s)
Aminopyridines/pharmacology , Interleukin-4/antagonists & inhibitors , Aminopyridines/metabolism , Humans , Interleukin-4/metabolism , Ligands , Phosphorylation/drug effects , Protein Binding , STAT6 Transcription Factor/chemistry , STAT6 Transcription Factor/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , THP-1 Cells
6.
Adv Drug Deliv Rev ; 144: 148-161, 2019 04.
Article in English | MEDLINE | ID: mdl-31491445

ABSTRACT

The foreign body response is an immunological process that leads to the rejection of implanted devices and presents a fundamental challenge to their performance, durability, and therapeutic utility. Recent advances in materials development and device design are now providing strategies to overcome this immune-mediated reaction. Here, we briefly review our current mechanistic understanding of the foreign body response and highlight new anti-FBR technologies from this decade that have been applied successfully in biomedical applications relevant to implants, devices, and cell-based therapies. Further development of these important technologies promises to enable new therapies, diagnostics, and revolutionize the management of patient care for many intractable diseases.


Subject(s)
Foreign-Body Reaction , Animals , Equipment and Supplies , Humans , Prostheses and Implants
7.
Biochemistry ; 58(4): 214-233, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30608114

ABSTRACT

The clinical onset of type 1 diabetes is characterized by the destruction of the insulin-producing ß cells of the pancreas and is caused by autoantigen-induced inflammation (insulitis) of the islets of Langerhans. The current standard of care for type 1 diabetes mellitus patients allows for management of the disease with exogenous insulin, but patients eventually succumb to many chronic complications such as limb amputation, blindness, and kidney failure. New therapeutic approaches now on the horizon are looking beyond glycemic management and are evaluating new strategies from protecting and regenerating endogenous islets to treating the underlying autoimmunity through selective modulation of key immune cell populations. Currently, there are no effective treatments for the autoimmunity that causes the disease, and strategies that aim to delay or prevent the onset of the disease will play an important role in the future of diabetes research. In this review, we summarize many of the key efforts underway that utilize molecular approaches to selectively modulate this disease and look at new therapeutic paradigms that can transform clinical treatment.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Hypoglycemic Agents/pharmacology , Immunologic Factors/pharmacology , Animals , Antigen-Presenting Cells/immunology , Clinical Trials as Topic , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Diabetes Mellitus, Type 1/pathology , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/immunology , Immunity, Innate , Immunomodulation/drug effects , Immunotherapy/methods , Insulin-Secreting Cells/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/physiopathology
8.
Nat Biomed Eng ; 2(11): 810-821, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30873298

ABSTRACT

The transplantation of pancreatic islet cells could restore glycaemic control in patients with type-I diabetes. Microspheres for islet encapsulation have enabled long-term glycaemic control in diabetic rodent models; yet human patients transplanted with equivalent microsphere formulations have experienced only transient islet-graft function, owing to a vigorous foreign-body reaction (FBR), to pericapsular fibrotic overgrowth (PFO) and, in upright bipedal species, to the sedimentation of the microspheres within the peritoneal cavity. Here, we report the results of the testing, in non-human primate (NHP) models, of seven alginate formulations that were efficacious in rodents, including three that led to transient islet-graft function in clinical trials. Although one month post-implantation all formulations elicited significant FBR and PFO, three chemically modified, immune-modulating alginate formulations elicited reduced FBR. In conjunction with a minimally invasive transplantation technique into the bursa omentalis of NHPs, the most promising chemically modified alginate derivative (Z1-Y15) protected viable and glucose-responsive allogeneic islets for 4 months without the need for immunosuppression. Chemically modified alginate formulations may enable the long-term transplantation of islets for the correction of insulin deficiency.

9.
Nat Mater ; 16(6): 671-680, 2017 06.
Article in English | MEDLINE | ID: mdl-28319612

ABSTRACT

Host recognition and immune-mediated foreign body response to biomaterials can compromise the performance of implanted medical devices. To identify key cell and cytokine targets, here we perform in-depth systems analysis of innate and adaptive immune system responses to implanted biomaterials in rodents and non-human primates. While macrophages are indispensable to the fibrotic cascade, surprisingly neutrophils and complement are not. Macrophages, via CXCL13, lead to downstream B cell recruitment, which further potentiated fibrosis, as confirmed by B cell knockout and CXCL13 neutralization. Interestingly, colony stimulating factor-1 receptor (CSF1R) is significantly increased following implantation of multiple biomaterial classes: ceramic, polymer and hydrogel. Its inhibition, like macrophage depletion, leads to complete loss of fibrosis, but spares other macrophage functions such as wound healing, reactive oxygen species production and phagocytosis. Our results indicate that targeting CSF1R may allow for a more selective method of fibrosis inhibition, and improve biomaterial biocompatibility without the need for broad immunosuppression.


Subject(s)
Biocompatible Materials/adverse effects , Foreign-Body Reaction/chemically induced , Foreign-Body Reaction/metabolism , Prostheses and Implants/adverse effects , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Foreign-Body Reaction/immunology , Mice , Primates
10.
Adv Healthc Mater ; 6(4)2017 Feb.
Article in English | MEDLINE | ID: mdl-27976536

ABSTRACT

The surface modification of implantable biomaterials with zwitterionic phosphorylcholine polymer is demonstrated through mussel-mimetic catecholamine polymer thin films. Using this method, the surfaces of alginate hydrogel microspheres and polystyrene microbeads, a model material known to produce robust foreign body responses and fibrosis, are successfully modified to reduce the tissue reaction by reducing the fibrosis in immunocompetent C57BL/6J mice.


Subject(s)
Catecholamines , Coated Materials, Biocompatible , Membranes, Artificial , Phosphorylcholine , Animals , Catecholamines/chemistry , Catecholamines/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Drug Implants/chemistry , Drug Implants/pharmacology , Fibrosis , Foreign-Body Reaction/prevention & control , Mice , Phosphorylcholine/chemistry , Phosphorylcholine/pharmacology
11.
Angew Chem Int Ed Engl ; 55(33): 9529-33, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27365192

ABSTRACT

The functionality of natural biopolymers has inspired significant effort to develop sequence-defined synthetic polymers for applications including molecular recognition, self-assembly, and catalysis. Conjugation of synthetic materials to biomacromolecules has played an increasingly important role in drug delivery and biomaterials. We developed a controlled synthesis of novel oligomers from hydroxyproline-based building blocks and conjugated these materials to siRNA. Hydroxyproline-based monomers enable the incorporation of broad structural diversity into defined polymer chains. Using a perfluorocarbon purification handle, we were able to purify diverse oligomers through a single solid-phase extraction method. The efficiency of synthesis was demonstrated by building 14 unique trimers and 4 hexamers from 6 diverse building blocks. We then adapted this method to the parallel synthesis of hundreds of materials in 96-well plates. This strategy provides a platform for the screening of libraries of modified biomolecules.


Subject(s)
Hydroxyproline/chemistry , Polyurethanes/chemical synthesis , Molecular Structure , Polyurethanes/chemistry , Solid Phase Extraction
14.
Nat Biotechnol ; 34(3): 345-52, 2016 03.
Article in English | MEDLINE | ID: mdl-26807527

ABSTRACT

The foreign body response is an immune-mediated reaction that can lead to the failure of implanted medical devices and discomfort for the recipient. There is a critical need for biomaterials that overcome this key challenge in the development of medical devices. Here we use a combinatorial approach for covalent chemical modification to generate a large library of variants of one of the most widely used hydrogel biomaterials, alginate. We evaluated the materials in vivo and identified three triazole-containing analogs that substantially reduce foreign body reactions in both rodents and, for at least 6 months, in non-human primates. The distribution of the triazole modification creates a unique hydrogel surface that inhibits recognition by macrophages and fibrous deposition. In addition to the utility of the compounds reported here, our approach may enable the discovery of other materials that mitigate the foreign body response.


Subject(s)
Foreign Bodies/immunology , Foreign-Body Reaction/immunology , Hydrogels/therapeutic use , Prostheses and Implants/adverse effects , Animals , Biocompatible Materials/adverse effects , Biocompatible Materials/therapeutic use , Humans , Hydrogels/adverse effects , Macrophages/immunology , Primates/immunology
15.
Nat Med ; 22(3): 306-11, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26808346

ABSTRACT

The transplantation of glucose-responsive, insulin-producing cells offers the potential for restoring glycemic control in individuals with diabetes. Pancreas transplantation and the infusion of cadaveric islets are currently implemented clinically, but these approaches are limited by the adverse effects of immunosuppressive therapy over the lifetime of the recipient and the limited supply of donor tissue. The latter concern may be addressed by recently described glucose-responsive mature beta cells that are derived from human embryonic stem cells (referred to as SC-ß cells), which may represent an unlimited source of human cells for pancreas replacement therapy. Strategies to address the immunosuppression concerns include immunoisolation of insulin-producing cells with porous biomaterials that function as an immune barrier. However, clinical implementation has been challenging because of host immune responses to the implant materials. Here we report the first long-term glycemic correction of a diabetic, immunocompetent animal model using human SC-ß cells. SC-ß cells were encapsulated with alginate derivatives capable of mitigating foreign-body responses in vivo and implanted into the intraperitoneal space of C57BL/6J mice treated with streptozotocin, which is an animal model for chemically induced type 1 diabetes. These implants induced glycemic correction without any immunosuppression until their removal at 174 d after implantation. Human C-peptide concentrations and in vivo glucose responsiveness demonstrated therapeutically relevant glycemic control. Implants retrieved after 174 d contained viable insulin-producing cells.


Subject(s)
Alginates , Blood Glucose/metabolism , C-Peptide/metabolism , Cell Transplantation/methods , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Embryonic Stem Cells/cytology , Foreign-Body Reaction/prevention & control , Hydrogels , Insulin-Secreting Cells/transplantation , Animals , Blotting, Western , Cell Culture Techniques , Cell Differentiation , Chromatography, Liquid , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunocompetence , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Mice , Microscopy, Confocal , Microscopy, Phase-Contrast , Morpholines , Polymers , Tandem Mass Spectrometry , Triazoles
16.
ACS Appl Mater Interfaces ; 8(3): 2255-61, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26713554

ABSTRACT

A high-throughput approach which automates the synthesis of polyelectrolyte-based layer-by-layer films (HT-LbL) to facilitate rapid film generation, systematic film characterization, and rational investigations into their interactions with cells is described. Key parameters, such as polyelectrolyte adsorption time and polyelectrolyte deposition pH, were used to modulate LbL film growth to create LbL films of distinct thicknesses using the widely utilized polyelectrolytes poly(allylamine hydrochloride) (PAH) and poly(acrylic acid) (PAA). We highlight how HT-LbL can be used to rapidly characterize film-forming parameters and robustly create linearly growing films of various molecular architectures. Film thickness and growth rates of HT-LbL films were shown to increase as a function of adsorption time. Subsequently, we investigated the role that polyelectrolyte solution pH (ranging from 2.5 to 9) has in forming molecularly distinct films of weak polyelectrolytes and report the effect this has on modulating cell attachment and spreading. Films synthesized at PAA-pH of 5.5 and PAH-pH 2.5-5.5 exhibited the highest cellular attachment. These results indicate that HT-LbL is a robust method that can shift the paradigm regarding the use of LbL in biomedical applications as it provides a rapid method to synthesize, characterize, and screen the interactions between molecularly distinct LbL films and cells.


Subject(s)
Electrolytes/pharmacology , Fibroblasts/cytology , Adsorption , Animals , Fibroblasts/drug effects , Hydrogen-Ion Concentration , Mice , NIH 3T3 Cells , Polymers/chemistry , Silicon/chemistry , Time Factors
17.
Nat Mater ; 14(6): 643-51, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25985456

ABSTRACT

The efficacy of implanted biomedical devices is often compromised by host recognition and subsequent foreign body responses. Here, we demonstrate the role of the geometry of implanted materials on their biocompatibility in vivo. In rodent and non-human primate animal models, implanted spheres 1.5 mm and above in diameter across a broad spectrum of materials, including hydrogels, ceramics, metals and plastics, significantly abrogated foreign body reactions and fibrosis when compared with smaller spheres. We also show that for encapsulated rat pancreatic islet cells transplanted into streptozotocin-treated diabetic C57BL/6 mice, islets prepared in 1.5-mm alginate capsules were able to restore blood-glucose control for up to 180 days, a period more than five times longer than for transplanted grafts encapsulated within conventionally sized 0.5-mm alginate capsules. Our findings suggest that the in vivo biocompatibility of biomedical devices can be significantly improved simply by tuning their spherical dimensions.


Subject(s)
Foreign-Body Reaction/immunology , Animals , Mice , Mice, Inbred C57BL , Primates
18.
Nat Rev Genet ; 15(8): 541-55, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25022906

ABSTRACT

Gene-based therapy is the intentional modulation of gene expression in specific cells to treat pathological conditions. This modulation is accomplished by introducing exogenous nucleic acids such as DNA, mRNA, small interfering RNA (siRNA), microRNA (miRNA) or antisense oligonucleotides. Given the large size and the negative charge of these macromolecules, their delivery is typically mediated by carriers or vectors. In this Review, we introduce the biological barriers to gene delivery in vivo and discuss recent advances in material sciences, nanotechnology and nucleic acid chemistry that have yielded promising non-viral delivery systems, some of which are currently undergoing testing in clinical trials. The diversity of these systems highlights the recent progress of gene-based therapy using non-viral approaches.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Animals , DNA/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Humans , RNA/genetics
19.
Nat Commun ; 5: 4277, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24969323

ABSTRACT

One of the most significant challenges in the development of clinically viable delivery systems for RNA interference therapeutics is to understand how molecular structures influence delivery efficacy. Here, we have synthesized 1,400 degradable lipidoids and evaluate their transfection ability and structure-function activity. We show that lipidoid nanoparticles mediate potent gene knockdown in hepatocytes and immune cell populations on IV administration to mice (siRNA EC50 values as low as 0.01 mg kg(-1)). We identify four necessary and sufficient structural and pKa criteria that robustly predict the ability of nanoparticles to mediate greater than 95% protein silencing in vivo. Because these efficacy criteria can be dictated through chemical design, this discovery could eliminate our dependence on time-consuming and expensive cell culture assays and animal testing. Herein, we identify promising degradable lipidoids and describe new design criteria that reliably predict in vivo siRNA delivery efficacy without any prior biological testing.


Subject(s)
Gene Knockdown Techniques/methods , Hepatocytes , Leukocytes , Lipids/chemistry , Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage , Animals , Drug Carriers , Mice , Transfection
20.
Proc Natl Acad Sci U S A ; 111(11): 3955-60, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24516150

ABSTRACT

siRNA therapeutics have promise for the treatment of a wide range of genetic disorders. Motivated by lipoproteins, we report lipopeptide nanoparticles as potent and selective siRNA carriers with a wide therapeutic index. Lead material cKK-E12 showed potent silencing effects in mice (ED50 ∼ 0.002 mg/kg), rats (ED50 < 0.01 mg/kg), and nonhuman primates (over 95% silencing at 0.3 mg/kg). Apolipoprotein E plays a significant role in the potency of cKK-E12 both in vitro and in vivo. cKK-E12 was highly selective toward liver parenchymal cell in vivo, with orders of magnitude lower doses needed to silence in hepatocytes compared with endothelial cells and immune cells in different organs. Toxicity studies showed that cKK-E12 was well tolerated in rats at a dose of 1 mg/kg (over 100-fold higher than the ED50). To our knowledge, this is the most efficacious and selective nonviral siRNA delivery system for gene silencing in hepatocytes reported to date.


Subject(s)
Drug Delivery Systems/methods , Lipopeptides/chemistry , Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage , Animals , Apolipoproteins E/metabolism , Cryoelectron Microscopy , Gene Silencing , Hepatocytes/metabolism , Macaca fascicularis , Mice , RNA, Small Interfering/therapeutic use , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...