Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Rep ; 14(1): 8938, 2024 04 18.
Article in English | MEDLINE | ID: mdl-38637629

ABSTRACT

Heart failure is a serious medical condition with a poor prognosis. Current treatments can only help manage the symptoms and slow the progression of heart failure. However, there is currently no cure to prevent and reverse cardiac remodeling. Transcription factors are in a central role in various cellular processes, and in the heart, GATA4 and NKX2-5 transcription factors mediate hypertrophic responses and remodeling. We have identified compounds that modulate the synergistic interaction of GATA4 and NKX2-5 and shown that the most promising compound (1, 3i-1000) is cardioprotective in vitro and in vivo. However, direct evidence of its binding site and mechanism of action has not been available. Due to the disordered nature of transcription factors, classical target engagement approaches cannot be utilized. Here, we synthesized a small-molecule ligand-binding pulldown probe of compound 1 to utilize affinity chromatography alongside CETSA, AlphaScreen, and molecular modeling to study ligand binding. These results provide the first evidence of direct physical binding of compound 1 selectively to GATA4. While developing drugs that target transcription factors presents challenges, advances in technologies and knowledge of intrinsically disordered proteins enable the identification of small molecules that can selectively target transcription factors.


Subject(s)
Heart Failure , Transcription Factors , Humans , Homeobox Protein Nkx-2.5/metabolism , Ligands , Transcription Factors/metabolism , Chromatography, Affinity , GATA4 Transcription Factor/metabolism , Homeodomain Proteins/metabolism
2.
Stem Cell Res Ther ; 15(1): 5, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167208

ABSTRACT

BACKGROUND: The prevalence of heart failure is constantly increasing, and the prognosis of patients remains poor. New treatment strategies to preserve cardiac function and limit cardiac hypertrophy are therefore urgently needed. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are increasingly used as an experimental platform for cardiac in vitro studies. However, in contrast to adult cardiomyocytes, hiPSC-CMs display immature morphology, contractility, gene expression and metabolism and hence express a naive phenotype that resembles more of a foetal cardiomyocyte. METHODS: A library of 14 novel compounds was synthesized in-house and screened for GATA4-NKX2-5 reporter activity and cellular toxicity. The most potent compound, 3i-1262, along with previously reported GATA4-acting compounds, were selected to investigate their effects on hypertrophy induced by endothelin-1 or mechanical stretch. Morphological changes and protein expression were characterized using immunofluorescence staining and high-content analysis. Changes in gene expression were studied using qPCR and RNA sequencing. RESULTS: The prototype compound 3i-1262 inhibited GATA4-NKX2-5 synergy in a luciferase reporter assay. Additionally, the isoxazole compound 3i-1262 inhibited the hypertrophy biomarker B-type natriuretic peptide (BNP) by reducing BNP promoter activity and proBNP expression in neonatal rat ventricular myocytes and hiPSC-CMs, respectively. Treatment with 3i-1262 increased metabolic activity and cardiac troponin T expression in hiPSC-CMs without affecting GATA4 protein levels. RNA sequencing analysis revealed that 3i-1262 induces gene expression related to metabolic activity and cell cycle exit, indicating a change in the identity and maturity status of hiPSC-CMs. The biological processes that were enriched in upregulated genes in response to 3i-1262 were downregulated in response to mechanical stretch, and conversely, the downregulated processes in response to 3i-1262 were upregulated in response to mechanical stretch. CONCLUSIONS: There is currently a lack of systematic understanding of the molecular modulation and control of hiPSC-CM maturation. In this study, we demonstrated that the GATA4-interfering compound 3i-1262 reorganizes the cardiac transcription factor network and converts hypertrophic signalling towards enhanced cardiomyocyte identity and maturity. This conceptually unique approach provides a novel structural scaffold for further development as a modality to promote cardiomyocyte specification and maturity.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Rats , Animals , Myocytes, Cardiac/metabolism , Induced Pluripotent Stem Cells/metabolism , Hypertrophy/metabolism , Transcription Factors/metabolism , Signal Transduction , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism
3.
Int J Mol Sci ; 22(24)2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34948382

ABSTRACT

Prior studies show that glycogen synthase kinase 3ß (GSK3ß) contributes to cardiac ischemic injury and cardiac hypertrophy. GSK3ß is constitutionally active and phosphorylation of GSK3ß at serine 9 (S9) inactivates the kinase and promotes cellular growth. GSK3ß is also phosphorylated at serine 389 (S389), but the significance of this phosphorylation in the heart is not known. We analyzed GSK3ß S389 phosphorylation in diseased hearts and utilized overexpression of GSK3ß carrying ser→ala mutations at S9 (S9A) and S389 (S389A) to study the biological function of constitutively active GSK3ß in primary cardiomyocytes. We found that phosphorylation of GSK3ß at S389 was increased in left ventricular samples from patients with dilated cardiomyopathy and ischemic cardiomyopathy, and in hearts of mice subjected to thoracic aortic constriction. Overexpression of either GSK3ß S9A or S389A reduced the viability of cardiomyocytes subjected to hypoxia-reoxygenation. Overexpression of double GSK3ß mutant (S9A/S389A) further reduced cardiomyocyte viability. Determination of protein synthesis showed that overexpression of GSK3ß S389A or GSK3ß S9A/S389A increased both basal and agonist-induced cardiomyocyte growth. Mechanistically, GSK3ß S389A mutation was associated with activation of mTOR complex 1 signaling. In conclusion, our data suggest that phosphorylation of GSK3ß at S389 enhances cardiomyocyte survival and protects from cardiomyocyte hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Myocardial Ischemia/metabolism , Myocytes, Cardiac/pathology , Animals , Cardiomegaly/pathology , Cell Proliferation , Cell Survival , Cells, Cultured , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Myocardial Ischemia/pathology , Myocytes, Cardiac/metabolism , Phosphorylation , Rats, Sprague-Dawley
4.
Stem Cell Res Ther ; 12(1): 190, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33736688

ABSTRACT

BACKGROUND: Pharmacological modulation of cell fate decisions and developmental gene regulatory networks holds promise for the treatment of heart failure. Compounds that target tissue-specific transcription factors could overcome non-specific effects of small molecules and lead to the regeneration of heart muscle following myocardial infarction. Due to cellular heterogeneity in the heart, the activation of gene programs representing specific atrial and ventricular cardiomyocyte subtypes would be highly desirable. Chemical compounds that modulate atrial and ventricular cell fate could be used to improve subtype-specific differentiation of endogenous or exogenously delivered progenitor cells in order to promote cardiac regeneration. METHODS: Transcription factor GATA4-targeted compounds that have previously shown in vivo efficacy in cardiac injury models were tested for stage-specific activation of atrial and ventricular reporter genes in differentiating pluripotent stem cells using a dual reporter assay. Chemically induced gene expression changes were characterized by qRT-PCR, global run-on sequencing (GRO-seq) and immunoblotting, and the network of cooperative proteins of GATA4 and NKX2-5 were further explored by the examination of the GATA4 and NKX2-5 interactome by BioID. Reporter gene assays were conducted to examine combinatorial effects of GATA-targeted compounds and bromodomain and extraterminal domain (BET) inhibition on chamber-specific gene expression. RESULTS: GATA4-targeted compounds 3i-1000 and 3i-1103 were identified as differential modulators of atrial and ventricular gene expression. More detailed structure-function analysis revealed a distinct subclass of GATA4/NKX2-5 inhibitory compounds with an acetyl lysine-like domain that contributed to ventricular cells (%Myl2-eGFP+). Additionally, BioID analysis indicated broad interaction between GATA4 and BET family of proteins, such as BRD4. This indicated the involvement of epigenetic modulators in the regulation of GATA-dependent transcription. In this line, reporter gene assays with combinatorial treatment of 3i-1000 and the BET bromodomain inhibitor (+)-JQ1 demonstrated the cooperative role of GATA4 and BRD4 in the modulation of chamber-specific cardiac gene expression. CONCLUSIONS: Collectively, these results indicate the potential for therapeutic alteration of cell fate decisions and pathological gene regulatory networks by GATA4-targeted compounds modulating chamber-specific transcriptional programs in multipotent cardiac progenitor cells and cardiomyocytes. The compound scaffolds described within this study could be used to develop regenerative strategies for myocardial regeneration.


Subject(s)
Nuclear Proteins , Transcription Factors , Cell Differentiation , Cell Line , GATA4 Transcription Factor/genetics , Homeobox Protein Nkx-2.5/genetics , Myocytes, Cardiac , Organogenesis , Transcription Factors/genetics
5.
Arch Toxicol ; 94(6): 2113-2130, 2020 06.
Article in English | MEDLINE | ID: mdl-32185414

ABSTRACT

Doxorubicin is a widely used anticancer drug that causes dose-related cardiotoxicity. The exact mechanisms of doxorubicin toxicity are still unclear, partly because most in vitro studies have evaluated the effects of short-term high-dose doxorubicin treatments. Here, we developed an in vitro model of long-term low-dose administration of doxorubicin utilizing human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Moreover, given that current strategies for prevention and management of doxorubicin-induced cardiotoxicity fail to prevent cancer patients developing heart failure, we also investigated whether the GATA4-targeted compound 3i-1000 has cardioprotective potential against doxorubicin toxicity both in vitro and in vivo. The final doxorubicin concentration used in the chronic toxicity model in vitro was chosen based on cell viability data evaluation. Exposure to doxorubicin at the concentrations of 1-3 µM markedly reduced (60%) hiPSC-CM viability already within 48 h, while a 14-day treatment with 100 nM doxorubicin concentration induced only a modest 26% reduction in hiPCS-CM viability. Doxorubicin treatment also decreased DNA content in hiPSC-CMs. Interestingly, the compound 3i-1000 attenuated doxorubicin-induced increase in pro-B-type natriuretic peptide (proBNP) expression and caspase-3/7 activation in hiPSC-CMs. Moreover, treatment with 3i-1000 for 2 weeks (30 mg/kg/day, i.p.) inhibited doxorubicin cardiotoxicity by restoring left ventricular ejection fraction and fractional shortening in chronic in vivo rat model. In conclusion, the results demonstrate that long-term exposure of hiPSC-CMs can be utilized as an in vitro model of delayed doxorubicin-induced toxicity and provide in vitro and in vivo evidence that targeting GATA4 may be an effective strategy to counteract doxorubicin-induced cardiotoxicity.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , GATA4 Transcription Factor/metabolism , Heart Diseases/prevention & control , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Protective Agents/pharmacology , Animals , Cardiotoxicity , Caspases, Effector/metabolism , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/metabolism , Protein Precursors/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Signal Transduction , Stroke Volume/drug effects , Time Factors , Ventricular Function, Left/drug effects
6.
J Med Chem ; 62(17): 8284-8310, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31431011

ABSTRACT

Transcription factors GATA4 and NKX2-5 directly interact and synergistically activate several cardiac genes and stretch-induced cardiomyocyte hypertrophy. Previously, we identified phenylisoxazole carboxamide 1 as a hit compound, which inhibited the GATA4-NKX2-5 transcriptional synergy. Here, the chemical space around the molecular structure of 1 was explored by synthesizing and characterizing 220 derivatives and structurally related compounds. In addition to the synergistic transcriptional activation, selected compounds were evaluated for their effects on transcriptional activities of GATA4 and NKX2-5 individually as well as potential cytotoxicity. The structure-activity relationship (SAR) analysis revealed that the aromatic isoxazole substituent in the southern part regulates the inhibition of GATA4-NKX2-5 transcriptional synergy. Moreover, inhibition of GATA4 transcriptional activity correlated with the reduced cell viability. In summary, comprehensive SAR analysis accompanied by data analysis successfully identified potent and selective inhibitors of GATA4-NKX2-5 transcriptional synergy and revealed structural features important for it.


Subject(s)
GATA4 Transcription Factor/antagonists & inhibitors , Homeobox Protein Nkx-2.5/antagonists & inhibitors , Isoxazoles/pharmacology , Animals , COS Cells , Cell Survival/drug effects , Cells, Cultured , Chlorocebus aethiops , Dose-Response Relationship, Drug , GATA4 Transcription Factor/chemistry , GATA4 Transcription Factor/metabolism , Homeobox Protein Nkx-2.5/chemistry , Homeobox Protein Nkx-2.5/metabolism , Isoxazoles/chemical synthesis , Isoxazoles/chemistry , Molecular Structure , Protein Binding/drug effects , Rats , Rats, Wistar , Structure-Activity Relationship
7.
Arch Toxicol ; 92(9): 2897-2911, 2018 09.
Article in English | MEDLINE | ID: mdl-29987409

ABSTRACT

Safety assessment of drug candidates in numerous in vitro and experimental animal models is expensive, time consuming and animal intensive. More thorough toxicity profiling already in the early drug discovery projects using human cell models, which more closely resemble the physiological cell types, would help to decrease drug development costs. In this study we aimed to compare different cardiac and stem cell models for in vitro toxicity testing and to elucidate structure-toxicity relationships of novel compounds targeting the cardiac transcription factor GATA4. By screening the effects of eight compounds at concentrations ranging from 10 nM up to 30 µM on the viability of eight different cell types, we identified significant cell type- and structure-dependent toxicity profiles. We further characterized two compounds in more detail using high-content analysis. The results highlight the importance of cell type selection for toxicity screening and indicate that stem cells represent the most sensitive screening model, which can detect toxicity that may otherwise remain unnoticed. Furthermore, our structure-toxicity analysis reveals a characteristic dihedral angle in the GATA4-targeted compounds that causes stem cell toxicity and thus helps to direct further drug development efforts towards non-toxic derivatives.


Subject(s)
GATA4 Transcription Factor/metabolism , High-Throughput Screening Assays/methods , Induced Pluripotent Stem Cells/drug effects , Small Molecule Libraries/toxicity , Toxicity Tests/methods , Animals , COS Cells , Cell Survival/drug effects , Cells, Cultured , Chlorocebus aethiops , Fibroblasts/drug effects , GATA4 Transcription Factor/genetics , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Myocytes, Cardiac/drug effects , Rats , Small Molecule Libraries/chemistry , Structure-Activity Relationship
8.
Sci Rep ; 8(1): 4611, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29545582

ABSTRACT

Transcription factors are fundamental regulators of gene transcription, and many diseases, such as heart diseases, are associated with deregulation of transcriptional networks. In the adult heart, zinc-finger transcription factor GATA4 is a critical regulator of cardiac repair and remodelling. Previous studies also suggest that NKX2-5 plays function role as a cofactor of GATA4. We have recently reported the identification of small molecules that either inhibit or enhance the GATA4-NKX2-5 transcriptional synergy. Here, we examined the cardiac actions of a potent inhibitor (3i-1000) of GATA4-NKX2-5 interaction in experimental models of myocardial ischemic injury and pressure overload. In mice after myocardial infarction, 3i-1000 significantly improved left ventricular ejection fraction and fractional shortening, and attenuated myocardial structural changes. The compound also improved cardiac function in an experimental model of angiotensin II -mediated hypertension in rats. Furthermore, the up-regulation of cardiac gene expression induced by myocardial infarction and ischemia reduced with treatment of 3i-1000 or when micro- and nanoparticles loaded with 3i-1000 were injected intramyocardially or intravenously, respectively. The compound inhibited stretch- and phenylephrine-induced hypertrophic response in neonatal rat cardiomyocytes. These results indicate significant potential for small molecules targeting GATA4-NKX2-5 interaction to promote myocardial repair after myocardial infarction and other cardiac injuries.


Subject(s)
GATA4 Transcription Factor/antagonists & inhibitors , Homeobox Protein Nkx-2.5/antagonists & inhibitors , Hypertension/prevention & control , Isoxazoles/pharmacology , Myocardial Infarction/prevention & control , Protein Interaction Domains and Motifs/drug effects , Reperfusion Injury/prevention & control , Small Molecule Libraries/pharmacology , Angiotensin II/toxicity , Animals , GATA4 Transcription Factor/metabolism , Gene Expression Regulation/drug effects , Homeobox Protein Nkx-2.5/metabolism , Hypertension/chemically induced , Hypertension/metabolism , Hypertension/pathology , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Phosphorylation , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
9.
J Med Chem ; 60(18): 7781-7798, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28858485

ABSTRACT

Transcription factors are pivotal regulators of gene transcription, and many diseases are associated with the deregulation of transcriptional networks. In the heart, the transcription factors GATA4 and NKX2-5 are required for cardiogenesis. GATA4 and NKX2-5 interact physically, and the activation of GATA4, in cooperation with NKX2-5, is essential for stretch-induced cardiomyocyte hypertrophy. Here, we report the identification of four small molecule families that either inhibit or enhance the GATA4-NKX2-5 transcriptional synergy. A fragment-based screening, reporter gene assay, and pharmacophore search were utilized for the small molecule screening, identification, and optimization. The compounds modulated the hypertrophic agonist-induced cardiac gene expression. The most potent hit compound, N-[4-(diethylamino)phenyl]-5-methyl-3-phenylisoxazole-4-carboxamide (3, IC50 = 3 µM), exhibited no activity on the protein kinases involved in the regulation of GATA4 phosphorylation. The identified and chemically and biologically characterized active compound, and its derivatives may provide a novel class of small molecules for modulating heart regeneration.


Subject(s)
GATA4 Transcription Factor/metabolism , Homeobox Protein Nkx-2.5/metabolism , Isoxazoles/chemistry , Isoxazoles/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Transcriptional Activation/drug effects , Animals , Cell Line , GATA4 Transcription Factor/agonists , GATA4 Transcription Factor/antagonists & inhibitors , Homeobox Protein Nkx-2.5/agonists , Homeobox Protein Nkx-2.5/antagonists & inhibitors , Humans , Mice , Models, Molecular , Protein Interaction Maps/drug effects
10.
Small ; 13(33)2017 09.
Article in English | MEDLINE | ID: mdl-28714245

ABSTRACT

Ischemic heart disease is the leading cause of death globally. Severe myocardial ischemia results in a massive loss of myocytes and acute myocardial infarction, the endocardium being the most vulnerable region. At present, current therapeutic lines only ameliorate modestly the quality of life of these patients. Here, an engineered nanocarrier is reported for targeted drug delivery into the endocardial layer of the left ventricle for cardiac repair. Biodegradable porous silicon (PSi) nanoparticles are functionalized with atrial natriuretic peptide (ANP), which is known to be expressed predominantly in the endocardium of the failing heart. The ANP-PSi nanoparticles exhibit improved colloidal stability and enhanced cellular interactions with cardiomyocytes and non-myocytes with minimal toxicity. After confirmation of good retention of the radioisotope 111-Indium in relevant physiological buffers over 4 h, in vivo single-photon emission computed tomography (SPECT/CT) imaging and autoradiography demonstrate increased accumulation of ANP-PSi nanoparticles in the ischemic heart, particularly in the endocardial layer of the left ventricle. Moreover, ANP-PSi nanoparticles loaded with a novel cardioprotective small molecule attenuate hypertrophic signaling in the endocardium, demonstrating cardioprotective potential. These results provide unique insights into the development of nanotherapies targeted to the injured region of the myocardium.


Subject(s)
Endocardium/pathology , Nanoparticles/chemistry , Signal Transduction , Animals , Cell Survival , Chemical Phenomena , Drug Delivery Systems , Drug Liberation , Hydrodynamics , Hypertrophy , Male , Nanoparticles/ultrastructure , Rats, Wistar , Surface Properties , Tissue Distribution
11.
Biomaterials ; 94: 93-104, 2016 07.
Article in English | MEDLINE | ID: mdl-27107168

ABSTRACT

Chronic heart failure, predominantly developed after myocardial infarction, is a leading cause of high mortality worldwide. As existing therapies have still limited success, natural and/or synthetic nanomaterials are emerging alternatives for the therapy of heart diseases. Therefore, we aimed to functionalize undecylenic acid thermally hydrocarbonized porous silicon nanoparticles (NPs) with different targeting peptides to improve the NP's accumulation in different cardiac cells (primary cardiomyocytes, non-myocytes, and H9c2 cardiomyoblasts), additionally to investigate the behavior of the heart-targeted NPs in vivo. The toxicity profiles of the NPs evaluated in the three heart-type cells showed low toxicity at concentrations up to 50 µg/mL. Qualitative and quantitative cellular uptake revealed a significant increase in the accumulation of atrial natriuretic peptide (ANP)-modified NPs in primary cardiomyocytes, non-myocytes and H9c2 cells, and in hypoxic primary cardiomyocytes and non-myocytes. Competitive uptake studies in primary cardiomyocytes showed the internalization of ANP-modified NPs takes place via the guanylate cyclase-A receptor. When a myocardial infarction rat model was induced by isoprenaline and the peptide-modified [(111)In]NPs administered intravenously, the targeting peptides, particularly peptide 2, improved the NPs' accumulation in the heart up to 3.0-fold, at 10 min. This study highlights the potential of these peptide-modified nanosystems for future applications in heart diseases.


Subject(s)
Heart/physiology , Nanoparticles/chemistry , Silicon/chemistry , Adsorption , Animals , Atrial Natriuretic Factor/metabolism , Blood Proteins/metabolism , Cell Survival , Colloids , Humans , Male , Myocytes, Cardiac/metabolism , Nanoparticles/ultrastructure , Peptides/chemistry , Porosity , Rats, Wistar , Temperature , Tomography, Emission-Computed, Single-Photon , Undecylenic Acids/chemistry
12.
PLoS One ; 10(12): e0144145, 2015.
Article in English | MEDLINE | ID: mdl-26642209

ABSTRACT

AIMS: Transcription factor GATA4 is a dosage sensitive regulator of heart development and alterations in its level or activity lead to congenital heart disease (CHD). GATA4 has also been implicated in cardiac regeneration and repair. GATA4 action involves combinatorial interaction with other cofactors such as NKX2-5, another critical cardiac regulator whose mutations also cause CHD. Despite its critical importance to the heart and its evolutionary conservation across species, the structural basis of the GATA4-NKX2-5 interaction remains incompletely understood. METHODS AND RESULTS: A homology model was constructed and used to identify surface amino acids important for the interaction of GATA4 and NKX2-5. These residues were subjected to site-directed mutagenesis, and the mutant proteins were characterized for their ability to bind DNA and to physically and functionally interact with NKX2-5. The studies identify 5 highly conserved amino acids in the second zinc finger (N272, R283, Q274, K299) and its C-terminal extension (R319) that are critical for physical and functional interaction with the third alpha helix of NKX2-5 homeodomain. Integration of the experimental data with computational modeling suggests that the structural arrangement of the zinc finger-homeodomain resembles the architecture of the conserved DNA binding domain of nuclear receptors. CONCLUSIONS: The results provide novel insight into the structural basis for protein-protein interactions between two important classes of transcription factors. The model proposed will help to elucidate the molecular basis for disease causing mutations in GATA4 and NKX2-5 and may be relevant to other members of the GATA and NK classes of transcription factors.


Subject(s)
DNA/metabolism , GATA4 Transcription Factor/metabolism , Heart Defects, Congenital/metabolism , Homeodomain Proteins/metabolism , Models, Biological , Mutation , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , COS Cells , Chlorocebus aethiops , DNA/genetics , GATA4 Transcription Factor/genetics , Heart Defects, Congenital/genetics , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Mice , Protein Binding , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Zinc Fingers
13.
Biomaterials ; 35(29): 8394-405, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24985734

ABSTRACT

Myocardial infarction (MI), commonly known as a heart attack, is the irreversible necrosis of heart muscle secondary to prolonged ischemia, which is an increasing problem in terms of morbidity, mortality and healthcare costs worldwide. Along with the idea to develop nanocarriers that efficiently deliver therapeutic agents to target the heart, in this study, we aimed to test the in vivo biocompatibility of different sizes of thermally hydrocarbonized porous silicon (THCPSi) microparticles and thermally oxidized porous silicon (TOPSi) micro and nanoparticles in the heart tissue. Despite the absence or low cytotoxicity, both particle types showed good in vivo biocompatibility, with no influence on hematological parameters and no considerable changes in cardiac function before and after MI. The local injection of THCPSi microparticles into the myocardium led to significant higher activation of inflammatory cytokine and fibrosis promoting genes compared to TOPSi micro and nanoparticles; however, both particles showed no significant effect on myocardial fibrosis at one week post-injection. Our results suggest that THCPSi and TOPSi micro and nanoparticles could be applied for cardiac delivery of therapeutic agents in the future, and the PSi biomaterials might serve as a promising platform for the specific treatment of heart diseases.


Subject(s)
Biocompatible Materials/chemistry , Drug Carriers/chemistry , Drug Delivery Systems , Myocardium/metabolism , Silicon/chemistry , Animals , Biocompatible Materials/adverse effects , Cells, Cultured , Drug Carriers/adverse effects , Fibrosis/chemically induced , Fibrosis/genetics , Gene Expression Regulation/drug effects , Inflammation/chemically induced , Inflammation/genetics , Male , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Porosity , Rats , Rats, Sprague-Dawley , Silicon/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...