Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Mol Ther ; 32(1): 227-240, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37925604

ABSTRACT

The novel severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), responsible for coronavirus disease 2019 (COVID-19), can trigger dysregulated immune responses known as the cytokine release syndrome (CRS), leading to severe organ dysfunction and respiratory distress. Our study focuses on developing an improved cell-permeable nuclear import inhibitor (iCP-NI), capable of blocking the nuclear transport of inflammation-associated transcription factors, specifically nuclear factor kappa B (NF-κB). By fusing advanced macromolecule transduction domains and nuclear localization sequences from human NF-κB, iCP-NI selectively interacts with importin α5, effectively reducing the expression of proinflammatory cytokines. In mouse models mimic SARS-CoV-2-induced pneumonitis, iCP-NI treatment demonstrated a significant decrease in mortality rates by suppressing proinflammatory cytokine production and immune cell infiltration in the lungs. Similarly, in hamsters infected with SARS-CoV-2, iCP-NI effectively protected the lung from inflammatory damage by reducing tumor necrosis factor-α, interleukin-6 (IL-6), and IL-17 levels. These promising results highlight the potential of iCP-NI as a therapeutic approach for COVID-19-related lung complications and other inflammatory lung diseases.


Subject(s)
COVID-19 , Mice , Animals , Humans , Transcription Factors/metabolism , Active Transport, Cell Nucleus , SARS-CoV-2 , NF-kappa B/metabolism , Inflammation , Cytokines/metabolism , Peptides/metabolism
2.
J Proteome Res ; 23(3): 985-998, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38306169

ABSTRACT

This study aims to elucidate the cellular mechanisms behind the secretion of complement factor B (CFB), known for its dual roles as an early biomarker for pancreatic ductal adenocarcinoma (PDAC) and as the initial substrate for the alternative complement pathway (ACP). Using parallel reaction monitoring analysis, we confirmed a consistent ∼2-fold increase in CFB expression in PDAC patients compared with that in both healthy donors (HD) and chronic pancreatitis (CP) patients. Elevated ACP activity was observed in CP and other benign conditions compared with that in HD and PDAC patients, suggesting a functional link between ACP and PDAC. Protein-protein interaction analyses involving key complement proteins and their regulatory factors were conducted using blood samples from PDAC patients and cultured cell lines. Our findings revealed a complex control system governing the ACP and its regulatory factors, including Kirsten rat sarcoma viral oncogene homolog (KRAS) mutation, adrenomedullin (AM), and complement factor H (CFH). Particularly, AM emerged as a crucial player in CFB secretion, activating CFH and promoting its predominant binding to C3b over CFB. Mechanistically, our data suggest that the KRAS mutation stimulates AM expression, enhancing CFH activity in the fluid phase through binding. This heightened AM-CFH interaction conferred greater affinity for C3b over CFB, potentially suppressing the ACP cascade. This sequence of events likely culminated in the preferential release of ductal CFB into plasma during the early stages of PDAC. (Data set ID PXD047043.).


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Complement Factor B/genetics , Complement Factor B/metabolism , Complement Pathway, Alternative , Proto-Oncogene Proteins p21(ras) , Early Detection of Cancer , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/genetics
3.
FASEB J ; 27(12): 4899-908, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23982142

ABSTRACT

Although thyroid-stimulating hormone (TSH) is known to be a major regulator of thyroid hormone biosynthesis and thyroid growth, insulin-like growth factor 1 (IGF-1) is required for mediating thyrocyte growth in concert with TSH in vitro. We generated mice with thyrocyte-selective ablation of IGF-1 receptor (TIGF1RKO) to explore the role of IGF-1 receptor signaling on thyroid function and growth. In 5-wk-old TIGF1RKO mice, serum thyroxine (T4) concentrations were decreased by 30% in concert with a 43% down-regulation of the monocarboxylate transporter 8 (MCT8), which is involved in T4 secretion. Despite a 3.5-fold increase in circulating concentrations of TSH, thyroid architecture and size were normal. Furthermore, thyrocyte area was increased by 40% in WT thyroids after 10 d TSH injection, but this effect was absent in TSH-injected TIGF1RKO mice. WT mice treated with methimazole and sodium perchlorate for 2 or 6 wk exhibited pronounced goiter development (2.0 and 5.4-fold, respectively), but in TIGF1RKO mice, goiter development was completely abrogated. These data reveal an essential role for IGF-1 receptor signaling in the regulation of thyroid function and TSH-stimulated goitrogenesis.


Subject(s)
Goiter/metabolism , Receptor, IGF Type 1/genetics , Thyrotropin/metabolism , Thyroxine/metabolism , Animals , Antithyroid Agents/pharmacology , Down-Regulation , Goiter/chemically induced , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Methimazole/pharmacology , Mice , Mice, Knockout , Monocarboxylic Acid Transporters , Perchlorates/toxicity , Receptor, IGF Type 1/deficiency , Sodium Compounds/toxicity , Symporters , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Gland/pathology
4.
Mater Today Bio ; 25: 100983, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38327977

ABSTRACT

The use of the FDA-approved osteoinductive growth factor BMP2 is widespread for bone regeneration. However, its clinical application has been hindered by limitations in cell permeability and a short half-life in circulation. To address this issue, we have developed a modified version of BMP2, referred to as Cell Permeable (CP)-BMP2, which possesses improved cell permeability. CP-BMP2 incorporates an advanced macromolecular transduction domain (aMTD) to facilitate transfer across the plasma membrane, a solubilization domain, and recombinant human BMP2. Compared to traditional rhBMP2, CP-BMP2 exhibits enhanced cell permeability, solubility, and bioavailability, and activates Smad phosphorylation through binding to BMP receptor 2. The effectiveness of CP-BMP2 was evaluated in three animal studies focusing on bone regeneration. In the initial study, mice and rabbits with critical-size calvarial defects received subcutaneous (SC) injections of CP-BMP2 and rhBMP2 (7.5 mg/kg, 3 injections per week for 8 weeks).Following 8 weeks of administration, CP-BMP2 demonstrated a remarkable 65 % increase in bone formation in mice when compared to both the vehicle and rhBMP2. Moreover, rabbits exhibited faster bone formation, characterized by a filling pattern originating from the center. In a subsequent study involving injured horses, hind limb bones treated with CP-BMP2 exhibited an 85 % higher bone regeneration rate, as evidenced by Micro-CT results, in contrast to horses treated with the vehicle or rhBMP2 (administered at 150 µg/defect, subcutaneously, once a week for 8 weeks, without a scaffold). These results underscore the potential of CP-BMP2 to facilitate rapid and effective healing. No noticeable adverse effects, such as ectopic bone formation, were observed in any of the studies. Overall, our findings demonstrate that CP-BMP2 holds therapeutic potential as a novel and effective osteogenic agent.

5.
Mol Ther ; 20(8): 1540-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22617107

ABSTRACT

Practical methods to deliver proteins systemically in animals have been hampered by poor tissue penetration and inefficient cytoplasmic localization of internalized proteins. We therefore pursued the development of improved macromolecule transduction domains (MTDs) and tested their ability to deliver therapeutically active p18(INK4c). MTD103 was identified from a screen of 1,500 signal peptides; tested for the ability to promote protein uptake by cells and tissues; and analyzed with regard to the mechanism of protein uptake and the delivery of biologically active p18(INK4c) into cancer cells. The therapeutic potential of cell-permeable MTD103p18(INK4c) (CP-p18(INK4c)) was tested in the HCT116 tumor xenograft model. MTD103p18(INK4c) appeared to traverse plasma membranes directly, was transferred from cell-to-cell and was therapeutically effective against cancer xenografts, inhibiting tumor growth by 86-98% after 5 weeks (P < 0.05). The therapeutic responses to CP-p18(INK4c) were accompanied by high levels of apoptosis in tumor cells. In addition to enhancing systemic delivery of CP-p18(INK4c) to normal tissues and cancer xenografts, the MTD103 sequence delayed protein clearance from the blood, liver and spleen. These results demonstrate that macromolecule intracellular transduction technology (MITT), enabled by MTDs, may provide novel protein therapies against cancer and other diseases.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinase Inhibitor p18/administration & dosage , Cyclin-Dependent Kinase Inhibitor p18/therapeutic use , Peptides/chemistry , Animals , Antineoplastic Agents/chemistry , Colorectal Neoplasms/drug therapy , Cyclin-Dependent Kinase Inhibitor p18/chemistry , Female , Humans , Mice , Mice, Inbred BALB C , Peptides/administration & dosage , Xenograft Model Antitumor Assays
6.
Nat Med ; 11(8): 892-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16007096

ABSTRACT

Suppressor of cytokine signaling (SOCS) 3 attenuates proinflammatory signaling mediated by the signal transducer and activator of transcription (STAT) family of proteins. But acute inflammation can occur after exposure to pathogen-derived inducers staphylococcal enterotoxin B (SEB) and lipopolysaccharide (LPS), or the lectin concanavalin A (ConA), suggesting that physiologic levels of SOCS3 are insufficient to stem proinflammatory signaling under pathogenic circumstances. To test this hypothesis, we developed recombinant cell-penetrating forms of SOCS3 (CP-SOCS3) for intracellular delivery to counteract SEB-, LPS- and ConA-induced inflammation. We found that CP-SOCS3 was distributed in multiple organs within 2 h and persisted for at least 8 h in leukocytes and lymphocytes. CP-SOCS3 protected animals from lethal effects of SEB and LPS by reducing production of inflammatory cytokines and attenuating liver apoptosis and hemorrhagic necrosis. It also reduced ConA-induced liver apoptosis. Thus, replenishing the intracellular stores of SOCS3 with CP-SOCS3 effectively suppresses the devastating effects of acute inflammation.


Subject(s)
Apoptosis/drug effects , Enterotoxins/toxicity , Inflammation/drug therapy , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Staphylococcal Infections/complications , Suppressor of Cytokine Signaling Proteins/therapeutic use , Animals , Concanavalin A/toxicity , Cytokines/blood , Inflammation/etiology , Leukocytes/metabolism , Lipopolysaccharides/toxicity , Liver/pathology , Lymphocytes/metabolism , Mice , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , STAT1 Transcription Factor/metabolism , Staphylococcal Infections/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Suppressor of Cytokine Signaling Proteins/pharmacokinetics , Suppressor of Cytokine Signaling Proteins/pharmacology
7.
Circ Res ; 103(11): 1259-69, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18849323

ABSTRACT

Diverse cardiac diseases induce cardiac hypertrophy, which leads to dilatation and heart failure. We previously reported that hypertrophy can be blocked by class I histone deacetylase (HDAC) inhibitor, which prompted us to investigate the regulatory mechanism of class I HDACs. Cardiac hypertrophy was introduced by aortic banding, by infusion of isoproterenol or angiotensin II, or by swimming. Hypertrophic stimuli transiently elevated the activity of histone deacetylase-2 (Hdac2), a class I HDAC. In cardiomyocytes, forced expression of Hdac2 simulated hypertrophy in an Akt-dependent manner, whereas enzymatically inert Hdac2 H141A failed to do so. Hypertrophic stimuli induced the expression of heat shock protein (Hsp)70. The induced Hsp70 physically associated with and activated Hdac2. Hsp70 overexpression produced a hypertrophic phenotype, which was blocked either by siHdac2 or by a dominant negative Hsp70DeltaABD. In Hsp70.1(-/-) mice, cardiac hypertrophy and Hdac2 activation were significantly blunted. Heat shock either to cardiomyocytes or to mice activated Hdac2 and induced hypertrophy. However, heat shock-induced Hdac2 activation was blunted in the cardiomyocytes isolated from Hsp70.1(-/-) mice. These results suggest that the induction of Hsp70 in response to diverse hypertrophic stresses and the ensuing activation of HDAC2 trigger cardiac hypertrophy, emphasizing HSP70/HDAC2 as a novel mechanism regulating hypertrophy.


Subject(s)
Cardiomegaly/physiopathology , Heat-Shock Response/physiology , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Animals , Cardiomegaly/enzymology , Cardiomegaly/genetics , Echocardiography , Enzyme Activation , Gene Expression Regulation , HSP72 Heat-Shock Proteins/deficiency , Histone Deacetylase 2 , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Rats , Transfection
8.
Sci Adv ; 6(18): eaba1193, 2020 05.
Article in English | MEDLINE | ID: mdl-32494688

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by mitochondrial dysfunction, Lewy body formation, and loss of dopaminergic neurons. Parkin, an E3 ubiquitin ligase, is thought to inhibit PD progression by removing damaged mitochondria and suppressing the accumulation of α-synuclein and other protein aggregates. The present study describes a protein-based therapy for PD enabled by the development of a cell-permeable Parkin protein (iCP-Parkin) with enhanced solubility and optimized intracellular delivery. iCP-Parkin recovered damaged mitochondria by promoting mitophagy and mitochondrial biogenesis and suppressed toxic accumulations of α-synuclein in cells and animals. Last, iCP-Parkin prevented and reversed declines in tyrosine hydroxylase and dopamine expression concomitant with improved motor function induced by mitochondrial poisons or enforced α-synuclein expression. These results point to common, therapeutically tractable features in PD pathophysiology, and suggest that motor deficits in PD may be reversed, thus providing opportunities for therapeutic intervention after the onset of motor symptoms.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Dopaminergic Neurons/metabolism , Mitochondria/metabolism , Parkinson Disease/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/genetics
9.
Endocrinology ; 157(1): 336-45, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26469138

ABSTRACT

IGF-1 receptor (IGF-1R) signaling is implicated in cardiac hypertrophy and longevity. However, the role of IGF-1R in age-related cardiac remodeling is only partially understood. We therefore sought to determine whether the deletion of the IGF-1R in cardiomyocytes might delay the development of aging-associated myocardial pathologies by examining 2-year-old male cardiomyocyte-specific IGF-1R knockout (CIGF1RKO) mice. Aging was associated with the induction of IGF-1R expression in hearts. Cardiomyocytes hypertrophied with age in wild-type (WT) mice. In contrast, the cardiac hypertrophic response associated with aging was blunted in CIGF1RKO mice. Concomitantly, fibrosis was reduced in aged CIGF1RKO compared with aged WT hearts. Expression of proinflammatory cytokines such as IL-1α, IL-1ß, IL-6, and receptor activator of nuclear factor-κB ligand was increased in aged WT hearts, but this increase was attenuated in aged CIGF1RKO hearts. Phosphorylation of Akt was increased in aged WT, but not in aged CIGF1RKO, hearts. In cultured cardiomyocytes, IGF-1 induced senescence as demonstrated by increased senescence-associated ß-galactosidase staining, and a phosphoinositide 3-kinase inhibitor inhibited this effect. Furthermore, inhibition of phosphoinositide 3-kinase significantly prevented the increase in IL-1α, IL-1ß, receptor activator of nuclear factor-κB ligand, and p21 protein expression by IGF-1. These data reveal an essential role for the IGF-1-IGF-1R-Akt pathway in mediating cardiomyocyte senescence.


Subject(s)
Aging , Cardiomegaly/metabolism , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Receptor, IGF Type 1/metabolism , Ventricular Remodeling , Animals , Biomarkers/metabolism , Cardiomegaly/immunology , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Cells, Cultured , Cellular Senescence/drug effects , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Fibrosis , Gene Expression Regulation, Developmental/drug effects , Heart Ventricles/drug effects , Heart Ventricles/immunology , Heart Ventricles/pathology , Insulin-Like Growth Factor I/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/agonists , Receptor, IGF Type 1/genetics , Signal Transduction/drug effects , Ventricular Remodeling/drug effects
10.
BMC Biotechnol ; 4: 25, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15500682

ABSTRACT

BACKGROUND: Cell-permeant Cre DNA site-specific recombinases provide an easily controlled means to regulate gene structure and function in living cells. Since recombination provides a stable and unambiguous record of protein uptake, the enzyme may also be used for quantitative studies of cis- and trans-acting factors that influence the delivery of proteins into cells. RESULTS: In the present study, 11 recombinant fusion proteins were analyzed to characterize sequences and conditions that affect protein uptake and/or activity and to develop more active cell-permeant enzymes. We report that the native enzyme has a low, but intrinsic ability to enter cells. The most active Cre proteins tested contained either an N-terminal 6xHis tag and a nuclear localization sequence from SV40 large T antigen (HNC) or the HIV Tat transduction sequence and a C-terminal 6xHis tag (TCH6). The NLS and 6xHis elements separately enhanced the delivery of the HNC protein into cells; moreover, transduction sequences from fibroblast growth factor 4, HIV Tat or consisting of the (KFF)3K sequence were not required for efficient protein transduction and adversely affected enzyme solubility. Transduction of the HNC protein required 10 to 15 min for half-maximum uptake, was greatly decreased at 4 degrees C and was inhibited by serum. Efficient recombination was observed in all cell types tested (a T-cell line, NIH3T3, Cos7, murine ES cells, and primary splenocytes), and did not require localization of the enzyme to the nucleus. CONCLUSIONS: The effects of different sequences on the delivery and/or activity of Cre in cultured cells could not be predicted in advance. Consequently, the process of developing more active cell-permeant recombinases was largely empirical. The HNC protein, with an excellent combination of activity, solubility and yield, will enhance the use of cell-permeant Cre proteins to regulate gene structure and function in living cells.


Subject(s)
Integrases/metabolism , Recombination, Genetic , Viral Proteins/metabolism , Animals , Cell Line , Cell Membrane Permeability , Cells, Cultured , Culture Media , Histidine/chemistry , Integrases/analysis , Integrases/genetics , Mice , Nuclear Localization Signals , Protein Transport , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Temperature , Viral Proteins/analysis , Viral Proteins/genetics
11.
Sci Rep ; 4: 4361, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24618595

ABSTRACT

The production of pluripotent stem cells (iPSCs) for therapeutic applications will require practical methods to achieve tight temporal and quantitative control of reprogramming factor (RF) expression, while avoiding the mutagenic potential of gene transfer. Toward this end, we have developed cell-permeable RF proteins (CP-RFs) incorporating newly developed macromolecule transduction domains (MTDs). Treatment of human dermal fibroblasts (HDFs) with combinations of cell-permeable OCT4, SOX2, KLF4, CMYC and either NANOG or LIN28 proteins induced the outgrowth of stem cell-like colonies (iSCs). iSC colonies generated with CP-RFs resembled embryonic stem cells with regard to morphology, biomarker expression, and extended capacity for self-renewal, but failed to expand as iPSC or ES cell lines. Partial reprogramming appears to be a common response to protein-based delivery of programming factors into somatic cells.


Subject(s)
Cell-Penetrating Peptides/metabolism , Cellular Reprogramming , Fibroblasts/metabolism , Recombinant Fusion Proteins/metabolism , Stem Cells/metabolism , Cell Differentiation , Cell Membrane Permeability , Cell-Penetrating Peptides/genetics , Cells, Cultured , Fibroblasts/cytology , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Stem Cells/cytology
12.
PLoS One ; 9(7): e102517, 2014.
Article in English | MEDLINE | ID: mdl-25019626

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder of complex etiology characterized by the selective loss of dopaminergic neurons, particularly in the substantia nigra. Parkin, a tightly regulated E3 ubiquitin ligase, promotes the survival of dopaminergic neurons in both PD and Parkinsonian syndromes induced by acute exposures to neurotoxic agents. The present study assessed the potential of cell-permeable parkin (CP-Parkin) as a neuroprotective agent. Cellular uptake and tissue penetration of recombinant, enzymatically active parkin was markedly enhanced by the addition of a hydrophobic macromolecule transduction domain (MTD). The resulting CP-Parkin proteins (HPM13 and PM10) suppressed dopaminergic neuronal toxicity in cells and mice exposed to 6-hydroxydopamine (6-OHDH) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). These included enhanced survival and dopamine expression in cultured CATH.a and SH-SY5Y neuronal cells; and protection against MPTP-induced damage in mice, notably preservation of tyrosine hydroxylase-positive cells with enhanced dopamine expression in the striatum and midbrain, and preservation of gross motor function. These results demonstrate that CP-Parkin proteins can compensate for intrinsic limitations in the parkin response and provide a therapeutic strategy to augment parkin activity in vivo.


Subject(s)
Neuroprotective Agents/pharmacology , Recombinant Proteins/pharmacology , Ubiquitin-Protein Ligases/pharmacology , Animals , Apoptosis , Cell Line , Cells, Cultured , Corpus Striatum/metabolism , Dopamine/biosynthesis , Dopamine/metabolism , Female , Hydrophobic and Hydrophilic Interactions , Mesencephalon/metabolism , Mice , Mice, Inbred BALB C , Motor Activity , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phenotype , Recombinant Proteins/chemistry , Tyrosine 3-Monooxygenase/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/physiology
13.
Clin Cancer Res ; 19(3): 680-90, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23230322

ABSTRACT

PURPOSE: Gastric cancer is a leading cause of cancer death worldwide. Limited therapeutic options highlight the need to understand the molecular changes responsible for the disease and to develop therapies based on this understanding. The goal of this study was to develop cell-permeable (CP-) forms of the RUNT-related transcription factor 3, RUNX3-a candidate tumor suppressor implicated in gastric and other epithelial cancers-to study the therapeutic potential of RUNX3 in the treatment of gastric cancer. EXPERIMENTAL DESIGN: We developed novel macromolecule transduction domains (MTD) which were tested for the ability to promote protein uptake by mammalian cells and tissues and used to deliver of biologically active RUNX3 into human gastric cancer cells. The therapeutic potential CP-RUNX3 was tested in the NCI-N87 human tumor xenograft animal model. RESULTS: RUNX3 fusion proteins, HM(57)R and HM(85)R, containing hydrophobic MTDs enter gastric cancer cells and suppress cell phenotypes (e.g., cell-cycle progression, wounded monolayer healing, and survival) and induce changes in biomarker expression (e.g., p21(Waf1) and VEGF) consistent with previously described effects of RUNX3 on TGF-ß signaling. CP-RUNX3 also suppressed the growth of subcutaneous human gastric tumor xenografts. The therapeutic response was comparable with studies augmenting RUNX3 gene expression in tumor cell lines; however, the protein was most active when administered locally, rather than systemically (i.e., intravenously). CONCLUSIONS: These results provide further evidence that RUNX3 can function as a tumor suppressor and suggest that practical methods to augment RUNX3 function could be useful in treating of some types of gastric cancer.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , Stomach Neoplasms/metabolism , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line , Core Binding Factor Alpha 3 Subunit/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Permeability , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins , Stomach Neoplasms/genetics , Xenograft Model Antitumor Assays
14.
Biomaterials ; 34(26): 6261-71, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23714245

ABSTRACT

Endostatin (ES), a 20 kDa protein derived from the carboxy-terminus of collagen XVIII is a potent angiogenesis inhibitor, but clinical development has been hindered by poor clinical efficacy and insufficient functional information from which to design agents with improved activity. The present study investigated protein uptake by cells as a determinant of ES activity. We developed a cell-permeable ES protein (HM73ES) with enhanced capacity to enter cells by adding a macromolecule transduction domain (MTD). HM73ES inhibited angiogenesis-associated phenotypes in cultured endothelial cells [as assessed by tube formation, wound-healing, cell proliferation and survival assays]. These effects were accompanied by reductions in MAPK signaling (ERK phosphorylation), and in ß-Catenin, c-Myc, STAT3, and VEGF protein expression. The cell-permeable ES displayed greater tissue penetration in mice and suppressed the growth of human tumor xenografts to a significantly greater extent than ES protein without the MTD sequence. Our results suggest that anti-angiogenic activities of native ES are limited at the level of protein uptake and/or subcellular localization, and that much of the activity of ES against tumors depends on one or more intracellular functions. This study will inform future efforts to understand ES function(s) and suggest strategies for improving ES-based cancer therapeutics.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Endostatins/pharmacokinetics , Endostatins/therapeutic use , Neoplasms/drug therapy , Amino Acid Sequence , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/genetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Endostatins/chemistry , Endostatins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , NIH 3T3 Cells , Neoplasms/blood supply , Neoplasms/pathology , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use
15.
Cancer Res ; 71(23): 7216-25, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21987726

ABSTRACT

Occult metastases are a major cause of cancer mortality, even among patients undergoing curative resection. Therefore, practical strategies to target the growth and persistence of already established metastases would provide an important advance in cancer treatment. Here, we assessed the potential of protein therapy using a cell permeable NM23-H1 metastasis suppressor protein. Hydrophobic transduction domains developed from a screen of 1,500 signaling peptide sequences enhanced the uptake of the NM23 protein by cultured cells and systemic delivery to animal tissues. The cell-permeable (CP)-NM23 inhibited metastasis-associated phenotypes in tumor cell lines, blocked the establishment of lung metastases, and cleared already established pulmonary metastases, significantly prolonging the survival of tumor-bearing animals. Therefore, these results establish the potential use of cell-permeable metastasis suppressors as adjuvant therapy against disseminated cancers.


Subject(s)
Lung Neoplasms/drug therapy , NM23 Nucleoside Diphosphate Kinases/pharmacology , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/pharmacokinetics , Cell-Penetrating Peptides/pharmacology , Disease Progression , Female , HCT116 Cells , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Sequence Data , Molecular Targeted Therapy/methods , NIH 3T3 Cells , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/pharmacokinetics , Neoplasm Metastasis , Protein Sorting Signals , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Xenograft Model Antitumor Assays
16.
Nat Neurosci ; 13(4): 482-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20190743

ABSTRACT

Fear can be acquired vicariously through social observation of others suffering from aversive stimuli. We found that mice (observers) developed freezing behavior by observing other mice (demonstrators) receive repetitive foot shocks. Observers had higher fear responses when demonstrators were socially related to themselves, such as siblings or mating partners. Inactivation of anterior cingulate cortex (ACC) and parafascicular or mediodorsal thalamic nuclei, which comprise the medial pain system representing pain affection, substantially impaired this observational fear learning, whereas inactivation of sensory thalamic nuclei had no effect. The ACC neuronal activities were increased and synchronized with those of the lateral amygdala at theta rhythm frequency during this learning. Furthermore, an ACC-limited deletion of Ca(v)1.2 Ca(2+) channels in mice impaired observational fear learning and reduced behavioral pain responses. These results demonstrate the functional involvement of the affective pain system and Ca(v)1.2 channels of the ACC in observational social fear.


Subject(s)
Calcium Channels, L-Type/physiology , Fear/physiology , Gyrus Cinguli/physiology , Learning/physiology , Pain/physiopathology , Social Behavior , Animals , Calcium Channels, L-Type/deficiency , Calcium Channels, L-Type/genetics , Cerebral Cortex/physiology , Conditioning, Classical/physiology , Fear/psychology , Female , Freezing Reaction, Cataleptic/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain/genetics , Pain/psychology , Pain Measurement/methods , Protein Subunits/deficiency , Protein Subunits/genetics , Protein Subunits/physiology , Rats , Rats, Sprague-Dawley
17.
J Cell Biochem ; 89(4): 674-87, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12858334

ABSTRACT

Protein transduction has been widely used to analyze biochemical processes in living cells quantitatively and under non-steady-state conditions. The present study analyzed the effects of cell cycle on the uptake and activity of cell-permeant Cre recombinase proteins. Previous studies had suggested that the efficiency of recombination and/or protein transduction varied among individual cells, even within a clonal population. We report here that cells in the G1 phase of the cell cycle undergo recombination at a lower rate than cells at other phases of the cell cycle, and that this variation results largely from differences in protein uptake, associated with differences in cell size. These results have implications regarding the mechanism of protein transduction and identify a source of heterogeneity that can influence the response of individual cells to cell-permeant proteins.


Subject(s)
Cell Cycle/physiology , Integrases/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Biological Transport , Blotting, Southern , Cell Cycle/genetics , Cell Membrane Permeability/genetics , Cell Membrane Permeability/physiology , Cell Size/physiology , Cells, Cultured , DNA, Bacterial/analysis , Escherichia coli/cytology , Escherichia coli/genetics , Escherichia coli/metabolism , Flow Cytometry , Genes, Reporter , Green Fluorescent Proteins , Integrases/drug effects , Integrases/genetics , Luminescent Proteins/metabolism , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombination, Genetic/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Viral Proteins/drug effects , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL