Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Biomater Sci ; 11(13): 4675-4683, 2023 Jun 27.
Article in English | MEDLINE | ID: mdl-37219049

ABSTRACT

Toxicity towards non-tumor cells during anticancer therapy can be reduced by using nanoscale systems for anticancer drug delivery. Usually only the loaded drug has anticancer activity. Recently, micellar nanocomplexes (MNCs) comprising green tea catechin derivatives for the delivery of the anticancer proteins, such as Herceptin, have been developed. Herceptin as well as the MNCs without the drug were effective against HER2/neu-overexpressing human tumor cells and had synergistic anticancer effects in vitro and in vivo. It remained unclear which kinds of negative effects the MNCs had on tumor cells exactly, and which of their components mediated them. Also, it was unclear if MNC has any toxicity effects on the normal cells of vital human organ systems. Herein we examined the effects of Herceptin-MNCs and their individual components on human breast cancer cells and on normal primary human endothelial and kidney proximal tubular cells. We applied a novel in vitro model that predicts nephrotoxicity in humans with high accuracy, as well as high-content screening and microfluidic mono- and co-culture models to thoroughly address effects on various cell types. The results showed that MNCs alone were profoundly toxic for breast cancer cells, and induced apoptosis regardless of HER2/neu expression levels. Apoptosis was induced by both green tea catechin derivatives contained within MNCs. In contrast, MNCs were not toxic for normal human cells, and the probability was low that MNCs would be nephrotoxic in humans. Together, the results supported the hypothesis that green tea catechin derivative-based MNCs could improve efficacy and safety of therapies with anticancer proteins.


Subject(s)
Breast Neoplasms , Catechin , Humans , Female , Micelles , Trastuzumab , Tea
2.
Biomater Sci ; 8(1): 497-505, 2019 Dec 17.
Article in English | MEDLINE | ID: mdl-31761907

ABSTRACT

Acute myeloid leukemia (AML) is an aggressive malignancy that leads to a poor prognosis even with intensive chemotherapy. As the key feature of AML is the blockade of hematopoietic cell maturation, considerable attention has been paid to 'differentiation therapy' aimed at transforming AML cells into more mature, benign phenotypes using pharmacological agents. Here we report a hyaluronic acid-(-)-epigallocatechin-3-O-gallate (HA-EGCG) conjugate as a unique anti-leukemic agent, capable of selectively killing AML cells as well as promoting their terminal differentiation into monocytes and granulocytes. This 'two-pronged' effect of the HA-EGCG conjugate was demonstrated in two different AML cell lines (NB4 and HL60), but absent in a physical mixture (HA + EGCG), highlighting the importance of HA conjugation for targeting of EGCG moieties to AML cells. Moreover, administration of the HA-EGCG conjugate not only suppressed AML progression, but also prolonged survival in the HL60 xenograft mouse model. Our study suggests new opportunities for designing two-pronged anti-leukemic agents for more effective AML treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Catechin/analogs & derivatives , Hyaluronic Acid/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Catechin/administration & dosage , Catechin/chemistry , Catechin/pharmacology , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , HEK293 Cells , HL-60 Cells , Human Umbilical Vein Endothelial Cells , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Male , Mice , Xenograft Model Antitumor Assays
3.
ACS Nano ; 13(7): 7591-7602, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31262169

ABSTRACT

Although a few nanomedicines have been approved for clinical use in cancer treatment, that recognizes improved patient safety through targeted delivery, their improved efficacy over conventional drugs has remained marginal. One of the typical drawbacks of nanocarriers for cancer therapy is a low drug-loading capacity that leads to insufficient efficacy and requires an increase in dosage and/or frequency of administration, which in turn increases carrier toxicity. In contrast, elevating drug-loading would cause the risk of nanocarrier instability, resulting in low efficacy and off-target toxicity. This intractable drug-to-carrier ratio has imposed constraints on the design and development of nanocarriers. However, if the nanocarrier has intrinsic therapeutic effects, the efficacy would be synergistically augmented with less concern for the drug-to-carrier ratio. Sunitinib-loaded micellar nanocomplex (SU-MNC) was formed using poly(ethylene glycol)-conjugated epigallocatechin-3-O-gallate (PEG-EGCG) as such a carrier. SU-MNC specifically inhibited the vascular endothelial growth factor-induced proliferation of endothelial cells, exhibiting minimal cytotoxicity to normal renal cells. SU-MNC showed enhanced anticancer effects and less toxicity than SU administered orally/intravenously on human renal cell carcinoma-xenografted mice, demonstrating more efficient effects on anti-angiogenesis, apoptosis induction, and proliferation inhibition against tumors. In comparison, a conventional nanocarrier, SU-loaded polymeric micelle (SU-PM) comprised of PEG-b-poly(lactic acid) (PEG-PLA) copolymer, only reduced toxicity with no elevated efficacy, despite comparable drug-loading and tumor-targeting efficiency to SU-MNC. Improved efficacy of SU-MNC was ascribed to the carrier-drug synergies with the high-performance carrier of PEG-EGCG besides tumor-targeted delivery.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Drug Delivery Systems , Kidney Neoplasms/drug therapy , Nanoparticles/chemistry , Sunitinib/pharmacology , Tea/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Catechin/analogs & derivatives , Catechin/chemistry , Cell Proliferation/drug effects , Cells, Cultured , Drug Carriers/chemistry , Female , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Nude , Mice, Transgenic , Micelles , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Particle Size , Polyethylene Glycols/chemistry , Sunitinib/administration & dosage , Sunitinib/chemistry , Surface Properties , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
4.
Adv Mater ; 30(14): e1706963, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29473233

ABSTRACT

Low drug loading and instability in blood circulation are two key challenges that impede the successful clinical translation of nanomedicine, as they result in only marginal therapeutic efficacy and toxic side effects associated with premature drug leakage, respectively. Herein, highly stable and ultrahigh drug loading micellar nanocomplexes (MNCs) based on the self-assembly of the anticancer drug doxorubicin (DOX) and a poly(ethylene glycol)-epigallocatechin-3-O-gallate (EGCG) conjugate are developed. The formation of these MNCs is facilitated by strong favorable intermolecular interactions between the structurally similar aromatic EGCG and DOX molecules, which impart exceptionally high drug-loading capability of up to 88% and excellent thermodynamic and kinetic stability. Unlike two clinical formulations of DOX-free DOX and liposomal DOX, which are not effective below their lethal dosages, these DOX-loaded MNCs demonstrate significant tumor growth inhibition in vivo on a human liver cancer xenograft mouse model with minimal unwanted toxicity. Overall, these MNCs can represent a safe and effective strategy to deliver DOX for cancer therapy.


Subject(s)
Nanostructures , Animals , Catechin , Cell Line, Tumor , Doxorubicin , Humans , Mice , Micelles , Neoplasms , Polyethylene Glycols , Tea
5.
Biomaterials ; 148: 41-53, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28961534

ABSTRACT

The green tea catechin, (-)-epigallocatechin-3-O-gallate (EGCG), has gained significant attention as a potent adjuvant to enhance the antitumor efficacy of cisplatin while mitigating its harmful side effects. Herein we report the development of a fail-safe cisplatin nanomedicine constructed with hyaluronic acid-EGCG conjugate for ovarian cancer therapy. A simple mixing of this conjugate and cisplatin induces spontaneous self-assembly of micellar nanocomplexes having a spherical core-shell structure. The surface-exposed hyaluronic acid enables efficient delivery of cisplatin into CD44-overexpressing cancer cells via receptor-mediated endocytosis whereas the internally packed EGCG moieties offer an environment favorable for the encapsulation of cisplatin. In addition, the antioxidant effect of EGCG moieties ensures fail-safe protection against off-target organ toxicity originating from cisplatin-evoked oxidative stress. Pharmacokinetic and biodistribution studies reveal the prolonged blood circulation and preferential tumor accumulation of intravenously administered nanocomplexes. Moreover, the nanocomplexes exhibit superior antitumor efficacy over free cisplatin while displaying no toxicity in both a subcutaneous xenograft model and peritoneal metastatic model of human ovarian cancer. Our findings demonstrate proof of concept for the feasibility of green tea catechin-based micellar nanocomplexes as a safe and effective cisplatin nanomedicine for ovarian cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Catechin/analogs & derivatives , Cisplatin/chemistry , Hyaluronic Acid/pharmacology , Nanoconjugates/chemistry , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Catechin/chemistry , Catechin/metabolism , Catechin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Liberation , Female , Humans , Hyaluronic Acid/chemistry , Mice, SCID , Micelles , Oxidative Stress/drug effects , Particle Size , Surface Properties , Tea/chemistry , Tissue Distribution
6.
Biomacromolecules ; 18(10): 3143-3155, 2017 Oct 09.
Article in English | MEDLINE | ID: mdl-28806066

ABSTRACT

Hyaluronic acid (HA)-based biomaterials have demonstrated only limited in vivo stability as a result of rapid degradation by hyaluronidase and reactive oxidative species. The green tea catechin, (-)-epigallocatechin-3-O-gallate (EGCG), has received considerable attention because of its powerful antioxidant and enzyme-inhibitory activities. We describe here the synthesis of HA-EGCG conjugate using a thiol-mediated reaction and its use for the preparation of a long-lasting injectable hydrogel. HA-EGCG conjugates with tunable degrees of substitution were synthesized by the nucleophilic addition reaction between EGCG quinone and thiolated HA under mild conditions. Contrary to unmodified HA, the conjugates exhibited free radical scavenging and hyaluronidase-inhibitory activities. Peroxidase-catalyzed coupling reaction between EGCG moieties was employed to produce in situ forming HA-EGCG hydrogel with surprisingly high resistance to hyaluronidase-mediated degradation. When injected subcutaneously in mice, HA-EGCG hydrogel was retained much longer than HA-tyramine hydrogel with minimal inflammation.


Subject(s)
Catechin/analogs & derivatives , Free Radical Scavengers/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Animals , Catechin/chemistry , Cell Line , Female , Free Radical Scavengers/adverse effects , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/pharmacokinetics , Hydrogels/adverse effects , Hydrogels/chemical synthesis , Hydrogels/pharmacokinetics , Macrophages/drug effects , Mice , Sulfhydryl Compounds/chemistry , Tissue Distribution
7.
J Control Release ; 226: 205-16, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-26855049

ABSTRACT

Nanosized polyelectrolyte complexes are attractive delivery vehicles for the transfer of therapeutic genes to diseased cells. Here we report the application of self-assembled ternary complexes constructed with plasmid DNA, branched polyethylenimine and hyaluronic acid-green tea catechin conjugates for targeted gene delivery. These conjugates not only stabilize plasmid DNA/polyethylenimine complexes via the strong DNA-binding affinity of green tea catechin, but also facilitate their transport into CD44-overexpressing cells via receptor-mediated endocytosis. The hydrodynamic size, surface charge and physical stability of the complexes are characterized. We demonstrate that the stabilized ternary complexes display enhanced resistance to nuclease attack and polyanion-induced dissociation. Moreover, the ternary complexes can efficiently transfect the difficult-to-transfect HCT-116 colon cancer cell line even in serum-supplemented media due to their enhanced stability and CD44-targeting ability. Confocal microscopic analysis demonstrates that the stabilized ternary complexes are able to promote the nuclear transport of plasmid DNA more effectively than binary complexes and hyaluronic acid-coated ternary complexes. The present study suggests that the ternary complexes stabilized with hyaluronic acid-green tea catechin conjugates can be widely utilized for CD44-targeted delivery of nucleic acid-based therapeutics.


Subject(s)
Catechin/analogs & derivatives , DNA/administration & dosage , Hyaluronic Acid/metabolism , Plasmids/administration & dosage , Transfection/methods , Catechin/chemistry , Catechin/metabolism , DNA/genetics , Endocytosis , Green Fluorescent Proteins/genetics , HCT116 Cells , HEK293 Cells , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Plasmids/genetics , Polyethyleneimine/chemistry , Polyethyleneimine/metabolism , Tea/chemistry , Tea/metabolism
8.
Acta Biomater ; 33: 142-52, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26785145

ABSTRACT

A novel ternary nanogel based on the self-assembly of hyaluronic acid-epigallocatechin gallate conjugates (HA-EGCG), linear polyethylenimine (PEI) and Granzyme B (GzmB) in an aqueous environment was developed for the targeted intracellular delivery of GzmB into cancer cells. Lysozyme-encapsulated HA-EGCG nanogels were first prepared and characterized. HA-EGCG nanogels exhibited smaller particle sizes and a more homogeneous size distribution than the HA counterpart. Fluorescence quenching and lysozyme activity studies revealed that EGCG moieties facilitated protein binding through physical interactions and led to the formation of stable nanogels. When CD44-overexpressing HCT-116 colon cancer cells were treated with GzmB-encapsulated HA-EGCG nanogels in vitro, a significant cytotoxic effect was observed. Caspase assays and intracellular trafficking studies confirmed that cell death was due to apoptosis triggered by the delivery of GzmB to the cytosol of those cells. In comparison, little cytotoxic effect was observed in CD44-deficient cells treated with GzmB-encapsulated HA-EGCG nanogels. This study highlights the potential utility of HA-EGCG as effective intracellular protein carriers for targeted cancer therapy. STATEMENT OF SIGNIFICANCE: Intracellularly activated cytotoxic proteins can be used to kill cancer cells but viable carriers for such proteins are lacking. In this work, we developed novel nanogels based on selfassembly of hyaluronic acid (HA)-(-)-epigallocatechin-3-gallate (EGCG) conjugates, linear polyethylenemine (PEI) and the cytotoxic protein Granzyme B (GzmB) for the intracellular delivery of GzmB for cancer therapy. HA was exploited for its ability to target CD44 which are overexpressed in many types of cancer cells, while EGCG, the main component of green tea catechins, was chosen for its ability to bind to proteins. Characterization studies showed that EGCG facilitated protein complexation through physical interactions and led to the formation of stable nanogels. HA-EGCG nanogels were able to achieve CD44 targeted killing of HCT-116 cancer cells by delivering GzmB into the cytosol of these cells. We believe that the applications of the HA-EGCG nanogels can be expanded to the intracellular delivery of other cytotoxic protein drugs for cancer therapy.


Subject(s)
Catechin/analogs & derivatives , Drug Delivery Systems/methods , Hyaluronic Acid/chemistry , Intracellular Space/metabolism , Muramidase/metabolism , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Tea/chemistry , Animals , Catechin/chemical synthesis , Catechin/chemistry , Cell Survival/drug effects , Chickens , Dimerization , Dynamic Light Scattering , Flow Cytometry , Granzymes/metabolism , HCT116 Cells , Hep G2 Cells , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemical synthesis , Nanogels , Spectrometry, Fluorescence
9.
J Mater Chem B ; 3(23): 4663-4670, 2015 Jun 21.
Article in English | MEDLINE | ID: mdl-32262481

ABSTRACT

A thermoresponsive injectable hydrogel scaffold for tissue engineering has been developed, whereby the scaffold was injected as a liquid at room temperature, and gelled at the target site in response to the change in body temperature. Our approach involved suspending thermoresponsive liposomes, which encapsulated horseradish peroxidase (HRP), in a hyaluronic acid-tyramine (HA-Tyr) conjugate and hydrogen peroxide (H2O2) solution. At room temperature, HRP was separated from the HA-Tyr conjugate by the lipid membrane, and hence the precursor solution remained as a liquid and was injectable. Upon injection and exposure to body temperature, the lipids experienced a phase transition, which significantly increased the membrane permeability and led to the release of HRP and the oxidative coupling of Tyr moieties with H2O2, forming a crosslinked hydrogel scaffold. It was shown in this study that the precursor solution remained as a liquid on the order of hours at 20 °C, yet gelation could be induced within minutes by heating to 37 °C. Furthermore, it was shown that HRP release could be controlled by various material and processing parameters, and that the gelation rate could be adjusted to meet various clinical needs by adjusting HRP encapsulation, liposome concentration and HA-Tyr concentration.

10.
ACS Macro Lett ; 4(9): 957-960, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-35596463

ABSTRACT

The oxidative coupling of phenols by horseradish peroxidase (HRP) is widely utilized to cross-link polymer-phenol conjugates for hydrogel formation. Phenols containing one aromatic ring are most commonly used, and the addition of hydrogen peroxide (H2O2) is an indispensable step in catalyzing the enzymatic reaction. We describe here a hydrogel composed of polyphenol as the cross-linking moiety. (-)-Epigallocatechin-3-gallate (EGCG), a green tea catechin, was conjugated to hyaluronic acid (HA) to form HA-EGCG conjugates. Addition of HRP to a solution of HA-EGCG conjugates at pH 7.4 induced gelation in 7 min. Notably, the addition of exogenous H2O2 was not required, as H2O2 was generated via EGCG autoxidation. Moreover, cross-linking between HA-EGCG conjugates occurred in situ through EGCG quinone formation, even when no HRP was added. This approach of forming hydrogels circumvented the safety concern associated with HRP due to its plant origin. Furthermore, the EGCG moieties endowed the hydrogels with resistance toward hyaluronidase-mediated degradation in vivo.

11.
Nat Nanotechnol ; 9(11): 907-912, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25282044

ABSTRACT

When designing drug carriers, the drug-to-carrier ratio is an important consideration, because the use of high quantities of carriers can result in toxicity as a consequence of poor metabolism and elimination of the carriers. However, these issues would be of less concern if both the drug and carrier had therapeutic effects. (-)-Epigallocatechin-3-O-gallate (EGCG), a major ingredient of green tea, has been shown, for example, to possess anticancer effects, anti-HIV effects, neuroprotective effects and DNA-protective effects. Here, we show that sequential self-assembly of the EGCG derivative with anticancer proteins leads to the formation of stable micellar nanocomplexes, which have greater anticancer effects in vitro and in vivo than the free protein. The micellar nanocomplex is obtained by complexation of oligomerized EGCG with the anticancer protein Herceptin to form the core, followed by complexation of poly(ethylene glycol)-EGCG to form the shell. When injected into mice, the Herceptin-loaded micellar nanocomplex demonstrates better tumour selectivity and growth reduction, as well as longer blood half-life, than free Herceptin.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Catechin/analogs & derivatives , Drug Carriers/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Catechin/chemistry , Catechin/therapeutic use , Cell Line, Tumor , Drug Carriers/chemistry , Humans , Mice , Mice, Nude , Micelles , Neoplasms/drug therapy , Neoplasms/pathology , Trastuzumab
12.
J Control Release ; 166(3): 203-10, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23328125

ABSTRACT

We report an injectable hydrogel system that incorporates interferon-α2a (IFN-α2a) for liver cancer therapy. IFN-α2a was incorporated in hydrogels composed of hyaluronic acid-tyramine (HA-Tyr) conjugates through the oxidative coupling of Tyr moieties with hydrogen peroxide (H2O2) and horseradish peroxidase (HRP). IFN-α2a-incorporated HA-Tyr hydrogels of varying stiffness were formed by changing the H2O2 concentration. The incorporation of IFN-α2a did not affect the rheological properties of the hydrogels. The activity of IFN-α2a was furthermore well-maintained in the hydrogels with lower stiffness. Through the caspase-3/7 pathway in vitro, IFN-α2a released from HA-Tyr hydrogels inhibited the proliferation of liver cancer cells and induced apoptosis. In the study of the pharmacokinetics, a higher concentration of IFN-α2a was shown in the plasma of mice treated with IFN-α2a-incorporated hydrogels after 4h post injection, with a much higher amount of IFN-α2a delivered at the tumor tissue comparing to that of injecting an IFN-α2a solution. The tumor regression study revealed that IFN-α2a-incorporated HA-Tyr hydrogels effectively inhibited tumor growth, while the injection of an IFN-α2a solution did not demonstrate antitumor efficacy. Histological studies confirmed that tumor tissues in mice treated with IFN-α2a-incorporated HA-Tyr hydrogels showed lower cell density, with more apoptotic and less proliferating cells compared with tissues treated with an IFN-α2a solution. In addition, the IFN-α2a-incorporated hydrogel treatment greatly inhibited the angiogenesis of tumor tissues.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Interferon-alpha/administration & dosage , Liver Neoplasms/drug therapy , Tyramine/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Carriers/chemical synthesis , Female , Humans , Hydrogels/chemistry , Injections, Subcutaneous , Interferon alpha-2 , Interferon-alpha/pharmacokinetics , Interferon-alpha/pharmacology , Interferon-alpha/therapeutic use , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Solubility , Xenograft Model Antitumor Assays
13.
Acta Biomater ; 8(5): 1826-37, 2012 May.
Article in English | MEDLINE | ID: mdl-22343003

ABSTRACT

An injectable hydrogel system, composed of gelatin-hydroxyphenylpropionic acid (Gtn-HPA) conjugates chemically cross-linked by an enzyme-mediated oxidation reaction, has been designed as a biodegradable scaffold for tissue engineering. In light of the role of substrate stiffness on cell differentiation, we herein report a newly improved Gtn hydrogel system with a broader range of stiffness control that uses Gtn-HPA-tyramine (Gtn-HPA-Tyr) conjugates to stimulate the osteogenic differentiation of human mesenchymal stem cells (hMSCs). The Gtn-HPA-Tyr conjugate was successfully synthesized through a further conjugation of Tyr to Gtn-HPA conjugate by means of a general carbodiimide/active ester-mediated coupling reaction. Proton nuclear magnetic resonance and UV-visible measurements showed a higher total phenol content in the Gtn-HPA-Tyr conjugate than that content in the Gtn-HPA conjugate. The Gtn-HPA-Tyr hydrogels were formed by the oxidative coupling of phenol moieties catalyzed by hydrogen peroxide (H(2)O(2)) and horseradish peroxidase (HRP). Rheological studies revealed that a broader range of storage modulus (G') of Gtn-HPA-Tyr hydrogel (600-26,800 Pa) was achieved using different concentrations of H(2)O(2), while the G' of the predecessor Gtn-HPA hydrogels was limited to the range of 1000 to 13,500 Pa. The hMSCs on Gtn-HPA-Tyr hydrogel with G' greater than 20,000 showed significantly up-regulated expressions of osteocalcin and runt-related transcription factor 2 (RUNX2) on both the gene and protein level, with the presence of alkaline phosphatase, and the evidence of calcium accumulation. These studies with the Gtn-HPA-Tyr hydrogel with G' greater than 20,000 collectively suggest the stimulation of the hMSCs into osteogenic differentiation, while these same observations were not found with the Gtn-HPA hydrogel with a G' of 13,500.


Subject(s)
Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteogenesis/physiology , Phenol/chemistry , Tissue Engineering/methods , Biocompatible Materials/chemical synthesis , Cell Differentiation , Cells, Cultured , Cross-Linking Reagents/chemistry , Elastic Modulus , Gelatin/chemistry , Humans , Materials Testing , Mesenchymal Stem Cells/physiology , Osteoblasts/physiology
14.
J Biomater Sci Polym Ed ; 23(14): 1793-806, 2012.
Article in English | MEDLINE | ID: mdl-21943785

ABSTRACT

We report an injectable hydrogel system with tunable stiffness for controlling the proliferation rate of human fibroblasts (HFF-1) in both two-dimensional (2D) and three-dimensional (3D) culture environments for potential use as a wound dressing material. The hydrogel composed of gelatin-hydroxyphenylpropionic acid (Gtn-HPA) conjugate was formed by the oxidative coupling of HPA moieties catalyzed by hydrogen peroxide (H2O2) and horseradish peroxidase (HRP). The stiffness of the hydrogels was controlled well by varying the H2O2 concentration. The effects of hydrogel stiffness on the proliferation rate of HFF-1 in both 2D and 3D were investigated. We found that the proliferation rate of HFF-1 using Gtn-HPA hydrogels was strongly dependent on the hydrogel stiffness, with a dimensionality-specific response. In the 2D studies, the HFF-1 exhibited a higher proliferation rate when the stiffness of the hydrogel was increased. In contrast, the HFF-1 cultured inside the hydrogel remained non-proliferative for 12 days before a stiffness-dependent proliferation profile was shown. The proliferation rate decreased with an increase in stiffness of the hydrogel in a 3D culture environment, unlike in a 2D environment.


Subject(s)
Cell Proliferation/physiology , Fibroblasts/physiology , Gelatin , Hydrogels , Actins/metabolism , Cell Adhesion , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Line , Elasticity , Fibroblasts/cytology , Gelatin/chemistry , Horseradish Peroxidase/chemistry , Humans , Hydrogels/chemical synthesis , Hydrogels/chemistry , Hydrogen Peroxide/chemistry , Microscopy, Confocal , Rheology , Water/chemistry
15.
Biomaterials ; 31(33): 8608-16, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20709390

ABSTRACT

We report the stimulation of neurogenesis and myogenesis of human mesenchymal stem cells (hMSCs) on the surfaces of biodegradable hydrogels with different stiffness. The hydrogels were composed of gelatin-hydroxyphenylpropionic acid (Gtn-HPA) conjugate were formed using the oxidative coupling of phenol moieties catalyzed by hydrogen peroxide (H(2)O(2)) and horseradish peroxidase (HRP). The storage modulus of the hydrogels was readily tuned from 600 to 12800 Pa. It was found that the stiffness of the hydrogel strongly affected the cell attachment, focal adhesion, migration and proliferation rate of hMSCs. The hMSCs on stiffer surfaces have a larger spreading area, more organized cytoskeletons, more stable focal adhesion, faster migration and a higher proliferation rate. The gene expression related to the extracellular matrix and adhesion molecules also differed when the cells were cultured on hydrogels with different stiffness. The differentiation of hMSCs on the surface of the hydrogel was closely linked to the hydrogel stiffness. The cells on a softer hydrogel (600 Pa) expressed more neurogenic protein markers, while cells on a stiffer hydrogel (12000 Pa) showed a higher up-regulation of myogenic protein markers.


Subject(s)
Cell Differentiation/drug effects , Collagenases/metabolism , Cross-Linking Reagents/pharmacology , Gelatin/chemistry , Gelatin/pharmacology , Hydrogels/chemistry , Materials Testing , Mesenchymal Stem Cells/cytology , Phenylpropionates/chemistry , Phenylpropionates/pharmacology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Hydrogels/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microscopy, Fluorescence , Rheology/drug effects
16.
Biomaterials ; 31(6): 1148-57, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19892395

ABSTRACT

We report an injectable hydrogel scaffold system with tunable stiffness for controlling the proliferation rate and differentiation of human mesenchymal stem cells (hMSCs) in a three-dimensional (3D) context in normal growth media. The hydrogels composed of gelatin-hydroxyphenylpropionic acid (Gtn-HPA) conjugate were formed using the oxidative coupling of HPA moieties catalyzed by hydrogen peroxide (H(2)O(2)) and horseradish peroxidase (HRP). The stiffness of the hydrogels was readily tuned by varying the H(2)O(2) concentration without changing the concentration of polymer precursor. We found that the hydrogel stiffness strongly affected the cell proliferation rates. The rate of hMSC proliferation increased with the decrease in the stiffness of the hydrogel. Also, the neurogenesis of hMSCs was controlled by the hydrogel stiffness in a 3D context without the use of any additional biochemical signal. These cells which were cultured in hydrogels with lower stiffness for 3 weeks expressed much more neuronal protein markers compared to those cultured within stiffer hydrogels for the same period of time.


Subject(s)
Absorbable Implants , Biocompatible Materials/chemistry , Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Nerve Regeneration/physiology , Neurons/cytology , Tissue Engineering/methods , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Crystallization/methods , Elasticity , Humans , Injections , Materials Testing , Mesenchymal Stem Cells/physiology , Neurons/physiology , Particle Size , Surface Properties , Viscosity
17.
J Control Release ; 134(3): 186-93, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19121348

ABSTRACT

Previously, we reported the independent tuning of mechanical strength (crosslinking density) and gelation rate of an injectable hydrogel system composed of hyaluronic acid-tyramine (HA-Tyr) conjugates. The hydrogels were formed through the oxidative coupling of tyramines which was catalyzed by hydrogen peroxide (H(2)O(2)) and horseradish peroxidase (HRP). Herein, we studied the encapsulation and release of model proteins using the HA-Tyr hydrogel. It was shown that the rapid gelation achieved by an optimal concentration of HRP could effectively encapsulate the proteins within the hydrogel network and thus prevented the undesired leakage of proteins into the surrounding tissues after injection. Hydrogels with different mechanical strengths were formed by changing the concentration of H(2)O(2) while maintaining the rapid gelation rate. The mechanical strength of the hydrogel controlled the release rate of proteins: stiff hydrogels released proteins slower compared to weak hydrogels. In phosphate buffer saline, alpha-amylase (negatively charged) was released sustainably from the hydrogel. Conversely, the release of lysozyme (positively charged) discontinued after the fourth hour due to electrostatic interactions with HA. In the presence of hyaluronidase, lysozymes were released continuously and completely from the hydrogel due to degradation of the hydrogel network. The activities of the released proteins were mostly retained which suggested that the HA-Tyr hydrogel is a suitable injectable and biodegradable system for the delivery of therapeutic proteins.


Subject(s)
Biocompatible Materials/chemistry , Cross-Linking Reagents/chemistry , Drug Carriers/chemistry , Hyaluronic Acid/analogs & derivatives , Hydrogels/chemistry , Proteins/administration & dosage , Tyramine/analogs & derivatives , Animals , Enzyme-Linked Immunosorbent Assay , Female , Hyaluronic Acid/chemistry , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Mice, Nude , Osmolar Concentration , Proteins/pharmacokinetics , Rheology , Tyramine/chemistry
18.
Soft Matter ; 4(4): 880-887, 2008 Mar 20.
Article in English | MEDLINE | ID: mdl-32907194

ABSTRACT

In this study, we propose an enzymatically crosslinked hyaluronic acid (HA) hydrogel with tunable mechanical strength and gelation rate as a novel injectable system. The hydrogel composed of HA-tyramine conjugate (HA-Tyr) was formed using the oxidative coupling of tyramine moieties catalyzed by hydrogen peroxide (H2O2) and horseradish peroxidase (HRP). The mechanical strength of the HA-Tyr hydrogel was tuned solely by the H2O2 amount without affecting the gelation rate. The hydrogels formed more rapidly with increasing HRP concentration and the gelation time ranged from 1 s to 20 min. A faster gelling system yielded more localized gel formation than a slower gelling one at the site where it was administered through subcutaneous injection. Studies on the swelling ratio and scanning electron microscopy images of the hydrogel structure further demonstrated that the crosslinking density was controlled by the concentration of H2O2 used. The mechanical strength of HA-Tyr hydrogels strongly affected the degradation rate in the presence of hyaluronidase in vitro; hydrogels degraded more slowly with increasing mechanical strength of the hydrogel. The independently tunable mechanical strength and gelation rate achieved by this enzymatically formed HA-Tyr hydrogel system will provide great advantages to a wide range of applications of injectable hydrogels, such as drug delivery and tissue regeneration.

19.
Biomaterials ; 28(3): 540-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16999995

ABSTRACT

Poor water solubility and low transfection efficiency of chitosan are major drawbacks for its use as a gene delivery carrier. PEGylation can increase its solubility, and folate conjugation may improve gene transfection efficiency due to promoted uptake of folate receptor-bearing tumor cells. The aim of this study was to synthesize and characterize folate-poly(ethylene glycol)-grafted chitosan (FA-PEG-Chi) for targeted plasmid DNA delivery to tumor cells. Gel electrophoresis study showed strong DNA binding ability of modified chitosan. The pH(50) values, defined as the pH when the transmittance of a polymer solution at 600 nm has reached 50% of the original value, suggested that the water solubility of PEGylated chitosan had improved significantly. Regression analysis of pH(50) value as a function of substitution degree of PEG yielded an almost linear correlation for PEG-Chi and FA-PEG-Chi. The solubility of PEGylated chitosan decreased slightly by further conjugation of folic acid due to the relatively more hydrophobic nature of folic acid when compared to PEG. In addition, the chitosan-based DNA complexes did not induce remarkable cytotoxicity against HEK 293 cells. FA-PEG-Chi can be a promising gene carrier due to its solubility in physiological pH, efficiency in condensing DNA, low cytotoxicity and targeting ability.


Subject(s)
Chitosan/chemistry , Folic Acid/chemistry , Gene Transfer Techniques , Polyethylene Glycols/chemistry , Carrier Proteins/metabolism , DNA/chemistry , Drug Carriers , Folate Receptors, GPI-Anchored , Genetic Vectors , Humans , Hydrogen-Ion Concentration , Models, Chemical , Neoplasms/therapy , Plasmids/metabolism , Receptors, Cell Surface/metabolism , Solubility , Water
20.
Chem Commun (Camb) ; (34): 4312-4, 2005 Sep 14.
Article in English | MEDLINE | ID: mdl-16113732

ABSTRACT

The sequential injection of hyaluronic acid-tyramine conjugates and enzymes forms biodegradable hydrogels in vivo by enzyme-induced oxidative coupling, offering high potential as a promising biomaterial for drug delivery and tissue engineering.


Subject(s)
Drug Delivery Systems , Hyaluronic Acid/chemistry , Hydrogels/chemical synthesis , Tissue Engineering , Tyramine/chemistry , Animals , Carbohydrate Sequence , Chemistry, Pharmaceutical , Horseradish Peroxidase/administration & dosage , Horseradish Peroxidase/pharmacokinetics , Hyaluronoglucosaminidase/chemistry , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...