Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Publication year range
1.
J Urol ; 195(3): 706-12, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26585682

ABSTRACT

PURPOSE: We compared levels of peroxiredoxin 2 in semen samples from normozoospermic and asthenozoospermic men. The potential effects of TAT-peroxiredoxin 2 fusion protein on sperm motility and DNA integrity were also evaluated. MATERIALS AND METHODS: Semen samples were obtained from 50 normozoospermic and 50 asthenozoospermic men. Lipid peroxidation of semen was determined using a commercial malondialdehyde kit. Sperm DNA fragmentation was evaluated by TUNEL assay. Western blot and immunofluorescence were performed to detect the amount of peroxiredoxin 2 protein in seminal plasma and spermatozoa. Sperm motility, DNA damage and levels of reactive oxygen species were evaluated after TAT-peroxiredoxin 2 fusion protein supplementation to the sperm suspension for 2 and 12 hours of incubation. RESULTS: In asthenozoospermic semen samples a significantly higher level of malondialdehyde and DNA damage was discovered. However, the expression of peroxiredoxin 2 was significantly lower in seminal plasma and spermatozoa compared with that of normozoospermic men. TAT-peroxiredoxin 2 fusion protein was successfully prepared and delivered to the spermatozoa. Interestingly adding TAT-peroxiredoxin 2 in asthenozoospermic sperm suspension effectively defended against the decrease in progressive motility and the increase in DNA damage. CONCLUSIONS: This study shows that supplementation of TAT-peroxiredoxin 2 fusion protein in the sperm suspension from asthenozoospermic men effectively improved sperm motility and DNA integrity by reducing levels of reactive oxygen species. Therefore, we speculate that peroxiredoxin 2 may have an important role as an antioxidant defense in semen and would provide new prevention and therapy alternatives for asthenozoospermia.


Subject(s)
Asthenozoospermia/drug therapy , Asthenozoospermia/genetics , DNA Damage/drug effects , Gene Products, tat/therapeutic use , Peroxiredoxins/analysis , Peroxiredoxins/therapeutic use , Semen/chemistry , Sperm Motility/drug effects , Spermatozoa/chemistry , Spermatozoa/drug effects , Adult , Asthenozoospermia/metabolism , Asthenozoospermia/physiopathology , Gene Products, tat/pharmacology , Humans , Male , Peroxiredoxins/pharmacology , Reactive Oxygen Species , Spermatozoa/physiology
2.
Exp Neurol ; 373: 114650, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38092186

ABSTRACT

Traumatic brain injury (TBI) is often associated with axonal injury that leads to significant motor and cognitive deficits. Ubiquitin carboxy terminal hydrolase L1 (UCHL1) is highly expressed in neurons and loss of its activity plays an important role in the pathogenesis of TBI. Fusion protein was constructed containing wild type (WT) UCHL1 and the HIV trans-activator of transcription capsid protein transduction domain (TAT-UCHL1) that facilitates transport of the protein into neurons after systemic administration. Additional mutant proteins bearing cysteine to alanine UCHL1 mutations at cysteine 152 (C152A TAT-UCHL1) that prevents nitric oxide and reactive lipid binding of C152, and at cysteine 220 (C220A TAT-UCHL1) that inhibits farnesylation of the C220 site were also constructed. WT, C152A, and C220A TAT-UCHL1 proteins administered to mice systemically after controlled cortical impact (CCI) were detectable in brain at 1 h, 4 h and 24 h after CCI by immunoblot. Mice treated with C152A or WT TAT-UCHL1 decreased axonal injury detected by NF200 immunohistochemistry 24 h after CCI, but C220A TAT-UCHL1 treatment had no significant effect. Further study indicated that WT TAT-UCHL1 treatment administered 24 h after CCI alleviated axonal injury as detected by SMI32 immunoreactivity 7 d after CCI, improved motor and cognitive deficits, reduced accumulation of total and K48-linked poly-Ub proteins, and attenuated the increase of the autophagy marker Beclin-1. These results suggest that UCHL1 activity contributes to the pathogenesis of white matter injury, and that restoration of UCHL1 activity by systemic treatment with WT TAT-UCHL1 after CCI may improve motor and cognitive deficits. These results also suggest that farnesylation of the C220 site may be required for the protective effects of UCHL1.


Subject(s)
Brain Injuries, Traumatic , Ubiquitin Thiolesterase , Mice , Animals , Ubiquitin Thiolesterase/genetics , Gene Products, tat/therapeutic use , Cysteine , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Axons/pathology
3.
Biochem Biophys Res Commun ; 417(3): 1024-9, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22222376

ABSTRACT

Chronic airway inflammation is a key feature of bronchial asthma. Annexin-1 (ANX1) is an anti-inflammatory protein that is an important modulator and plays a key role in inflammation. Although the precise action of ANX1 remains unclear, it has emerged as a potential drug target for inflammatory diseases such as asthma. To examine the protective effects of ANX1 protein on ovalbumin (OVA)-induced asthma in animal models, we used a cell-permeable Tat-ANX1 protein. Mice sensitized and challenged with OVA antigen had an increased amount of cytokines and eosinophils in their bronchoalveolar lavage (BAL) fluid. However, administration of Tat-ANX1 protein before OVA challenge significantly decreased the levels of cytokines (interleukin (IL)-4, IL-5, and IL-13) and BAL fluid in lung tissues. Furthermore, OVA significantly increased the activation of mitogen-activated protein kinase (MAPK) in lung tissues, whereas Tat-ANX1 protein markedly reduced phosphorylation of MAPKs such as extracellular signal-regulated protein kinase, p38, and stress-activated protein kinase/c-Jun N-terminal kinase. These results suggest that transduced Tat-ANX1 protein may be a potential protein therapeutic agent for the treatment of lung disorders including asthma.


Subject(s)
Annexin A1/therapeutic use , Annexins/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Asthma/drug therapy , Gene Products, tat/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Animals , Annexin A1/administration & dosage , Annexins/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Asthma/prevention & control , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Disease Models, Animal , Female , Gene Products, tat/administration & dosage , Mice , Mice, Inbred BALB C , Ovalbumin , Recombinant Fusion Proteins/administration & dosage
4.
Int J Biol Macromol ; 209(Pt A): 972-983, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35460749

ABSTRACT

Rett syndrome (RTT) is a neurodevelopmental disorder caused by pathogenic variants leading to functional impairment of the MeCP2 protein. Here, we used purified recombinant MeCP2e1 and MeCP2e2 protein variants fused to a TAT protein transduction domain (PTD) to evaluate their transduction ability into RTT patient-derived fibroblasts and the ability to carry out their cellular function. We then assessed their transduction ability and therapeutic effects in a RTT mouse model. In vitro, TAT-MeCP2e2-eGFP reversed the pathological hyperacetylation of histones H3K9 and H4K16, a hallmark of abolition of MeCP2 function. In vivo, intraperitoneal administration of TAT-MeCP2e1 and TAT-MeCP2e2 extended the lifespan of Mecp2-/y mice by >50%. This was accompanied by rescue of hippocampal CA2 neuron size in animals treated with TAT-MeCP2e1. Taken together, these findings provide a strong indication that recombinant TAT-MeCP2 can reach mouse brains following peripheral injection and can ameliorate the phenotype of RTT mouse models. Thus, our study serves as a first step in the development of a potentially novel RTT therapy.


Subject(s)
Rett Syndrome , Animals , Disease Models, Animal , Gene Products, tat/genetics , Gene Products, tat/therapeutic use , Humans , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mutation , Phenotype , Rett Syndrome/drug therapy , Rett Syndrome/genetics , Rett Syndrome/metabolism
5.
Chem Biol Interact ; 344: 109495, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-33961834

ABSTRACT

Cataracts, a clouding of the eye lens, are a leading cause of visual impairment and are responsible for one of the most commonly performed surgical procedures worldwide. Although generally safe and effective, cataract surgery can lead to a secondary lens abnormality due to transition of lens epithelial cells to a mesenchymal phenotype (EMT) and opacification of the posterior lens capsular bag. Occurring in up to 40% of cataract cases over time, posterior capsule opacification (PCO) introduces additional treatment costs and reduced quality of life for patients. Studies have shown that PCO pathogenesis is driven in part by TGF-ß, signaling through the action of the family of Smad coactivators to effect changes in gene transcription. In the present study, we evaluated the ability of Smad-7, a well characterized inhibitor of TGF-ß -mediated Smad signaling, to suppress the EMT response in lens epithelial cells associated with PCO pathogenesis. Treatment of lens epithelial cells with a cell-permeable form of Smad7 variant resulted in suppressed expression of EMT markers such as alpha smooth muscle actin and fibronectin. A single application of cell-permeable Smad7 variant in the capsular bag of a mouse cataract surgery model resulted in suppression of gene transcripts encoding alpha smooth muscle actin and fibronectin. These results point to Smad7 as a promising biotherapeutic agent for prevention or substantial reduction in the incidence of PCO following cataract surgery.


Subject(s)
Capsule Opacification/prevention & control , Cell-Penetrating Peptides/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Gene Products, tat/therapeutic use , Lens, Crystalline/drug effects , Smad7 Protein/therapeutic use , Actins/metabolism , Animals , Capsule Opacification/etiology , Capsule Opacification/pathology , Cataract/complications , Cataract/pathology , Epithelial Cells/drug effects , Lens, Crystalline/pathology , Mice, Transgenic , Protein Domains , Recombinant Proteins/therapeutic use
6.
Blood ; 112(6): 2474-83, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18574026

ABSTRACT

Nucleophosmin (NPM) is frequently overexpressed in leukemias and other tumors. NPM has been reported to suppress oncogene-induced senescence and apoptosis and may represent a therapeutic target for cancer. We fused a NPM-derived peptide to the HIV-TAT (TAT-NPMDeltaC) and found that the fusion peptide inhibited proliferation and induced apoptotic death of primary fibroblasts and preleukemic stem cells. TAT-NPMDeltaC down-regulated several NF-kappaB-controlled survival and inflammatory proteins and suppressed NF-kappaB-driven reporter gene activities. Using an inflammation-associated leukemia model, we demonstrate that TAT-NPMDeltaC induced proliferative suppression and apoptosis of preleukemic stem cells and significantly delayed leukemic development in mice. Mechanistically, TAT-NPMDeltaC associated with wild-type NPM proteins and formed complexes with endogenous NPM and p65 at promoters of several antiapoptotic and inflammatory genes and abrogated their transactivation by NF-kappaB in leu-kemic cells. Thus, TAT-delivered NPM peptide may provide a novel therapy for inflammation-associated tumors that require NF-kappaB signaling for survival.


Subject(s)
Apoptosis/drug effects , Gene Products, tat/therapeutic use , Leukemia/drug therapy , Nuclear Proteins/administration & dosage , Peptide Fragments/administration & dosage , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Inflammation , Leukemia/pathology , Mice , NF-kappa B/antagonists & inhibitors , Neoplastic Stem Cells/pathology , Nucleophosmin , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use
7.
Mol Cancer ; 8: 97, 2009 Nov 09.
Article in English | MEDLINE | ID: mdl-19900276

ABSTRACT

BACKGROUND: Based on its role in angiogenesis and apoptosis, the inhibition of NFkappaB activity is considered an effective treatment for cancer, hampered by the lack of selective and safe inhibitors. We recently demonstrated that the RH domain of GRK5 (GRK5-RH) inhibits NFkappaB, thus we evaluated its effects on cancer growth. METHODS: The role of GRK5-RH on tumor growth was assessed in a human cancer cell line (KAT-4). RH overexpression was induced by adenovirus mediated gene transfer; alternatively we administered a synthetic protein reproducing the RH domain of GRK5 (TAT-RH), actively transported into the cells. RESULTS: In vitro, adenovirus mediated GRK5-RH overexpression (AdGRK5-NT) in human tumor cells (KAT-4) induces IkappaB accumulation and inhibits NFkappaB transcriptional activity leading to apoptotic events. In BALB/c nude mice harboring KAT-4 induced neoplasias, intra-tumor delivery of AdGRK5-NT reduces in a dose-dependent fashion tumor growth, with the highest doses completely inhibiting it. This phenomenon is paralleled by a decrease of NFkappaB activity, an increase of IkappaB levels and apoptotic events. To move towards a pharmacological setup, we synthesized the TAT-RH protein. In cultured KAT-4 cells, different dosages of TAT-RH reduced cell survival and increased apoptosis. In BALB/c mice, the anti-proliferative effects of TAT-RH appear to be dose-dependent and highest dose completely inhibits tumor growth. CONCLUSION: Our data suggest that GRK5-RH inhibition of NFkappaB is a novel and effective anti-tumoral strategy and TAT-RH could be an useful tool in the fighting of cancer.


Subject(s)
Gene Products, tat/pharmacology , NF-kappa B/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Proteins/pharmacology , Adenoviridae/genetics , Animals , Annexin A5/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , G-Protein-Coupled Receptor Kinase 5/chemistry , G-Protein-Coupled Receptor Kinase 5/metabolism , Gene Products, tat/administration & dosage , Gene Products, tat/therapeutic use , Humans , Inflammation/complications , Inflammation/genetics , Mice , Mice, Inbred BALB C , Neoplasms/blood supply , Neoplasms/genetics , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/genetics , Protein Structure, Tertiary , Recombinant Proteins/administration & dosage , Recombinant Proteins/isolation & purification , Recombinant Proteins/therapeutic use , Staining and Labeling , Xenograft Model Antitumor Assays
8.
Neurobiol Dis ; 34(1): 87-94, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19167500

ABSTRACT

Cerebral ischemia activates endogenous neurogenesis in the subventricular zone (SVZ) and the dentate gyrus. Consecutively, SVZ-derived neural precursors migrate towards ischemic lesions. However, functional relevance of activated neurogenesis is limited by poor survival of new-born precursors. We therefore employed the HI-virus-derived fusion protein TAT-Bcl-x(L) to study the effects of acute anti-apoptotic treatment on endogenous neurogenesis and functional outcome after transient cerebral ischemia in mice. TAT-Bcl-x(L) treatment led to significantly reduced acute ischemic cell death (128+/-23 vs. 305+/-65 TUNEL+ cells/mm(2) in controls) and infarct volumes resulting in less motor deficits and improved spatial learning. It significantly increased survival of doublecortin (Dcx)-positive neuronal precursors (389+/-96 vs. 213+/-97 Dcx+ cells in controls) but did not enhance overall post-ischemic cell proliferation or lesion-specific neuronal differentiation 28 days after ischemia. Our data demonstrate that post-stroke TAT-Bcl-x(L)-treatment results in acute neuroprotection, improved functional outcome, and enhanced survival of lesion-specific neuronal precursor cells after cerebral ischemia in mice.


Subject(s)
Brain Ischemia/drug therapy , Corpus Striatum/physiopathology , Gene Products, tat/therapeutic use , Neuroprotective Agents/therapeutic use , Stem Cells/physiology , bcl-X Protein/therapeutic use , Animals , Brain Ischemia/physiopathology , Cell Death/physiology , Cell Survival/physiology , Doublecortin Domain Proteins , Doublecortin Protein , Learning , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Motor Activity , Neurogenesis/physiology , Neuropeptides/metabolism , Recombinant Fusion Proteins/therapeutic use , Space Perception
9.
ACS Chem Neurosci ; 10(1): 412-423, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30117729

ABSTRACT

Early brain injury (EBI) after aneurysmal subarachnoid hemorrhage (SAH) contributes to high morbidity and mortality. Although it is well recognized that acute neuroinflammation reaction is one of the most important triggers of EBI, pharmacotherapy proved to be clinically effective against the initiating of neuroinflammation after SAH is lacking. The resident microglia and infiltrated peripheral monocyte are two main types of immune cells in central nervous system (CNS) and control the inflammation process in brain after SAH. But the time course and relative contributions of these two immune cell activations after SAH are unknown. The p75 neurotrophin receptor (p75NTR), member of TNF receptor superfamily, expresses on infiltrated peripheral monocytes and suppresses their proinflammatory action after brain insults. But the p75NTR expression on resident microglia in vivo is rarely explored and their function keeps elusive. Therefore, we designed this study to investigate the time course of resident microglia activation and peripheral monocyte infiltration, as well as the microglial expression of p75NTR by using CX3C-chemokine receptor 1 (Cx3cr1) and chemokine receptor 2 (Ccr2) double transgenic mice (Cx3cr1GFP/+Ccr2RFP/+) after SAH. The results showed activated microglia was observed in cortex as early as 24 h and further increased at 48 and 72 h post SAH, while the infiltrated monocyte was not found until 72h. In addition, activated microglia expressed p75NTR acutely and p75NTR specific antagonist TAT-Pep5 significantly reduced microglia activation, neuroinflammation and EBI from 24 to 72 h. Together, these data suggest that the early neuroinflammation reaction might be initiated and intensified mainly by resident microglia rather than infiltrated monocyte at least in the first 48 h after SAH and p75NTR blockading by TAT-Pep5P might alleviate EBI through mediating microglial activation.


Subject(s)
Brain Injuries/metabolism , Microglia/metabolism , Monocytes/metabolism , Neuroprotective Agents/pharmacology , Receptors, Nerve Growth Factor/metabolism , Subarachnoid Hemorrhage/metabolism , Animals , Brain Injuries/etiology , Brain Injuries/prevention & control , Female , Gene Products, tat/pharmacology , Gene Products, tat/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Monocytes/drug effects , Neuroprotective Agents/therapeutic use , Random Allocation , Receptors, Nerve Growth Factor/antagonists & inhibitors , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy
10.
Biochem J ; 407(2): 285-92, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17627607

ABSTRACT

The use of CPPs (cell-penetrating peptides) as delivery vectors for bioactive molecules has been an emerging field since 1994 when the first CPP, penetratin, was discovered. Since then, several CPPs, including the widely used Tat (transactivator of transcription) peptide, have been developed and utilized to translocate a wide range of compounds across the plasma membrane of cells both in vivo and in vitro. Although the field has emerged as a possible future candidate for drug delivery, little attention has been given to the potential toxic side effects that these peptides might exhibit in cargo delivery. Also, no comprehensive study has been performed to evaluate the relative efficacy of single CPPs to convey different cargos. Therefore we selected three of the major CPPs, penetratin, Tat and transportan 10, and evaluated their ability to deliver commonly used cargos, including fluoresceinyl moiety, double-stranded DNA and proteins (i.e. avidin and streptavidin), and studied their effect on membrane integrity and cell viability. Our results demonstrate the unfeasibility to use the translocation efficacy of fluorescein moiety as a gauge for CPP efficiency, since the delivery properties are dependent on the cargo used. Furthermore, and no less importantly, the toxicity of CPPs depends heavily on peptide concentration, cargo molecule and coupling strategy.


Subject(s)
Drug Delivery Systems/adverse effects , Peptides/therapeutic use , Carrier Proteins/therapeutic use , Carrier Proteins/toxicity , Cell Membrane Permeability , Cell Survival/drug effects , Cell-Penetrating Peptides , DNA/administration & dosage , DNA/pharmacokinetics , Drug Delivery Systems/methods , Drug-Related Side Effects and Adverse Reactions , Galanin/therapeutic use , Galanin/toxicity , Gene Products, tat/therapeutic use , Gene Products, tat/toxicity , HeLa Cells , Humans , Peptides/toxicity , Proteins/administration & dosage , Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Recombinant Fusion Proteins/toxicity , Wasp Venoms/therapeutic use , Wasp Venoms/toxicity
11.
Oncogene ; 25(52): 6968-74, 2006 Nov 02.
Article in English | MEDLINE | ID: mdl-16702945

ABSTRACT

The inhibitor of apoptosis gene family member Survivin is highly expressed in most tumors, and appears to be a promising target for cancer therapy. Although a variety of Survivin antagonists have been shown to induce apoptosis in malignant cells, the potential utility of these agents is limited by inefficient delivery and cell impermeability. We generated recombinant fusion proteins containing the TAT protein transduction domain and either wild-type Survivin (TAT-Surv-WT) or a dominant-negative mutant (TAT-Surv-T34A). The TAT-Surv proteins were purified by sequential affinity and ion-exchange chromatography, and at 30 nM concentration demonstrated rapid entry into cells at 30 min. Whereas TAT-Surv-WT had minimal effect on YUSAC2 or WM793 melanoma cells, TAT-Surv-T34A induced cell detachment, DNA fragmentation, caspase-3 activation and mitochondrial release of apoptosis-inducing factor at low microM concentrations. Intraperitoneal (i.p.) injection of mice bearing subcutaneous YUSAC2 xenografts with TAT-Surv-T34A (10 mg/kg) was associated with rapid tumor accumulation at 1 h, and increased tumor cell apoptosis and aberrant nuclei formation in situ. Repeated i.p. injection of TAT-Surv-T34A resulted in a 40-50% reduction in growth and mass of established tumors, compared to those similarly injected with saline buffer or TAT-Surv-WT. These studies demonstrate the feasibility of systemic tumor treatment using a cell-permeable Survivin antagonist.


Subject(s)
Apoptosis/physiology , Gene Products, tat/therapeutic use , Melanoma/pathology , Melanoma/therapy , Microtubule-Associated Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation/drug effects , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Inhibitor of Apoptosis Proteins , Mice , Neoplasms, Experimental/therapy , Survivin , Transduction, Genetic
12.
Trends Pharmacol Sci ; 27(9): 455-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16876260

ABSTRACT

The 5-hydroxytryptamine (5-HT)(2C) receptor has received considerable attention as a target for treating drug addiction. 5-HT(2C)-receptor agonism, however, also induces side-effects. In this article, we review recent findings regarding the involvement of 5-HT(2C) receptors in behaviours related to drug addiction in animals. It was recently shown that 5-HT(2C)-receptor agonist effects can be induced intracellularly using the protein peptide Tat-3L4F, which prevents 5-HT(2C)-receptor dephosphorylation induced by phosphatase and tensin homologue deleted on chromosome 10. The most promising finding is that Tat-3L4F can selectively reduce the potency of addictive drugs by reducing mesolimbic dopamine transmission without eliciting the side-effects of 5-HT(2C)-receptor agonist treatment, thus highlighting its potential use as a strategy to treat drug addiction in humans.


Subject(s)
Gene Products, tat/therapeutic use , Peptide Fragments/therapeutic use , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin Receptor Agonists/therapeutic use , Substance-Related Disorders/drug therapy , Animals , Dopamine/metabolism , Humans , PTEN Phosphohydrolase/antagonists & inhibitors , Phosphorylation , Serotonin Receptor Agonists/adverse effects
13.
Peptides ; 87: 50-63, 2017 01.
Article in English | MEDLINE | ID: mdl-27887988

ABSTRACT

The failure of proteins to penetrate mammalian cells or target tumor cells restricts their value as therapeutic tools in a variety of diseases such as cancers. Recently, protein transduction domains (PTDs) or cell penetrating peptides (CPPs) have been shown to promote the delivery of therapeutic proteins or peptides into live cells. The successful delivery of proteins mainly depends on their physicochemical properties. Although, linear cell penetrating peptides are one of the most effective delivery vehicles; but currently, cyclic CPPs has been developed to potently transport bioactive full-length proteins into cells. Up to now, several small protein transduction domains from viral proteins including Tat or VP22 could be fused to other peptides or proteins to entry them in various cell types at a dose-dependent approach. A major disadvantage of PTD-fusion proteins is primary uptake into endosomal vesicles leading to inefficient release of the fusion proteins into the cytosol. Recently, non-covalent complex formation (Chariot) between proteins and CPPs has attracted a special interest to overcome some delivery limitations (e.g., toxicity). Many preclinical and clinical trials of CPP-based delivery are currently under evaluation. Generally, development of more efficient protein transduction domains would significantly increase the potency of protein therapeutics. Moreover, the synergistic or combined effects of CPPs with other delivery systems for protein/peptide drug delivery would promote their therapeutic effects in cancer and other diseases. In this review, we will describe the functions and implications of CPPs for delivering the therapeutic proteins or peptides in preclinical and clinical studies.


Subject(s)
Cell-Penetrating Peptides/therapeutic use , Drug Delivery Systems , Proteins/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Cell-Penetrating Peptides/chemistry , Endosomes/chemistry , Gene Products, tat/chemistry , Gene Products, tat/therapeutic use , Humans , Protein Domains/genetics , Proteins/chemistry
14.
Brain Res ; 1082(1): 61-6, 2006 Apr 12.
Article in English | MEDLINE | ID: mdl-16703672

ABSTRACT

Glial-cell-line-derived neurotrophic factor (GDNF) promotes mesencephalic dopaminergic neuronal survival in several in vitro and in vivo models. As the demise of dopaminergic neurons is the cause for Parkinson's disease (PD) symptoms, GDNF is a promising agent for its treatment. However, this neurotrophin is unable to cross the blood-brain barrier, which has complicated its clinical use. Therefore, ways to deliver GDNF into the central nervous system in an effective manner are needed. The HIV-1-Tat-derived cell-penetrating peptide (CPP) provides a means to deliver fusion proteins into the brain. We generated a fusion protein between the 11 amino acid CPP of Tat and the rat GDNF mature protein to deliver GDNF across the blood-brain barrier. We showed previously that Tat-GDNF enhances the neuroprotective effect of GDNF in in vivo models for nerve trauma and ischemia. Here, we tested its effect in a subchronic scheme of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) application into the mouse as a model for PD to evaluate the effect of Tat-GDNF fusion protein in dopaminergic neuron survival. We showed that the fusion protein did indeed reach the dopaminergic neurons. However, the in vivo application of Tat-GDNF did not provide neuroprotection of dopaminergic neurons, as revealed by immunohistochemistry and counting of the number of tyrosine-hydroxylase-immunoreactive neurons in the substantia nigra pars compacta. Possibly, GDNF does protect nigro-striatal projections of those neurons that survive MPTP treatment but does not increase the number of surviving dopaminergic neurons. A concomitant treatment of Tat-GDNF with an anti-apoptotic Tat-fusion protein might be beneficial.


Subject(s)
Blood-Brain Barrier/physiopathology , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Animals , Blood-Brain Barrier/drug effects , Cell Count/methods , Disease Models, Animal , Drug Interactions , Gene Products, tat/therapeutic use , Gene Transfer Techniques , Genes, tat , Genetic Vectors , Immunohistochemistry/methods , Lentivirus , Mice , Parkinson Disease/etiology , Peptides/pharmacology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
15.
Neurosci Lett ; 401(1-2): 40-3, 2006 Jun 19.
Article in English | MEDLINE | ID: mdl-16650576

ABSTRACT

Future gene therapy for brainstem variant amyotrophic lateral sclerosis may be technically difficult if gene therapy vectors are injected near vital cardiorespiratory centers or if large portions of the tongue and pharyngeal muscles must be peripherally injected for retrograde transport of vectors to motor neurons. In this study we show that it is possible to deliver recombinant proteins to the hypoglossal nuclei without brainstem or muscle injections, by taking advantage of enhanced uptake of fusion proteins containing the protein transduction domain from the human immunodeficiency virus TAT protein. Adenoviral vectors expressing either TAT-modified or native beta-glucuronidase (beta-gluc) were injected into the lateral cerebral ventricles of mice, transducing ventricular epithelium down to the level of the obex in the brainstem. There was significant uptake into the hypoglossal nuclei of TAT-modified but not native beta-glucuronidase. The TAT-modified beta-gluc appeared to encompass half or more of the hypoglossal nuclei as visualized by retrograde labeling with cholera toxin subunit b in adjacent sections. TAT-modification of gene products may allow a relatively non-invasive approach to brainstem gene therapy via cerebroventricular injection.


Subject(s)
Gene Products, tat/metabolism , Genetic Vectors/genetics , Hypoglossal Nerve/metabolism , Recombinant Fusion Proteins/metabolism , Transduction, Genetic/methods , Transfection/methods , Adenoviridae/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/therapy , Animals , Area Postrema/cytology , Area Postrema/metabolism , Area Postrema/virology , Ependyma/cytology , Ependyma/metabolism , Ependyma/virology , Fourth Ventricle/cytology , Fourth Ventricle/metabolism , Fourth Ventricle/virology , Gene Products, tat/genetics , Gene Products, tat/therapeutic use , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Glucuronidase/genetics , Hypoglossal Nerve/cytology , Hypoglossal Nerve/virology , Injections, Intraventricular , Medulla Oblongata/cytology , Medulla Oblongata/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/cytology , Motor Neurons/metabolism , Protein Structure, Tertiary/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Transduction, Genetic/trends , Transfection/trends
16.
Epilepsy Res ; 72(1): 75-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16930946

ABSTRACT

In this work we analysed the characteristics of the cell-permeable peptide TAT-PTD fused to cystatin B (CSTB) to evaluate its potential for protein therapy of Unverricht-Lundborg (UL) epilepsy. TAT-PTD-CSTB does not penetrate the cells despite initial evidence of time and concentration-dependent transduction. Therefore, it cannot be used as a form of replacement of the intracytoplasmic protein missing in UL. Importantly, we discuss precautions to avoid false-positive results when working with TAT-PTD for protein therapy of neurological diseases.


Subject(s)
Cystatins/metabolism , Gene Products, tat/metabolism , Transduction, Genetic , Unverricht-Lundborg Syndrome/therapy , Blood-Brain Barrier , Cell Membrane/physiology , Cystatin B , Cystatins/genetics , Cystatins/therapeutic use , Cysteine Proteinase Inhibitors , Gene Products, tat/genetics , Gene Products, tat/therapeutic use , Humans , Plasmids , Protein Binding , Protein Transport
17.
Oncogene ; 23(5): 1153-65, 2004 Feb 05.
Article in English | MEDLINE | ID: mdl-14691460

ABSTRACT

Apoptin has been described to induce apoptosis in various human cancer cell lines, but not in normal cells, thus making it an interesting candidate for the development of novel therapeutic strategies. Apoptin was generated and cloned into several mammalian expression vectors. Transfection or microinjection of apoptin cDNA resulted in its expression, initially in the cytoplasm with a filamentous pattern. Subsequently, apoptin entered the nucleus and efficiently induced apoptosis in several cancer cell lines. Nuclear localization was shown to be required for induction of apoptosis. Apoptin expression level was found to be an important determinant of the efficiency of induction of apoptosis. Surprisingly, expression of apoptin or GFP-apoptin cDNA induced apoptosis in some normal cells. When fused to the HIV-TAT protein transduction domain and delivered as a protein, TAT-apoptin was transduced efficiently (>90%) into normal and tumour cells. However, TAT-apoptin remained in the cytoplasm and did not kill normal 6689 and 1BR3 fibroblasts. In contrast TAT-apoptin migrated from the cytoplasm to the nucleus of Saos-2 and HSC-3 cancer cells resulting in apoptosis after 24 h. This study shows that apoptin is a powerful apoptosis-inducing protein with a potential for cancer therapy.


Subject(s)
Apoptosis , Capsid Proteins/therapeutic use , Gene Products, tat/therapeutic use , Genetic Therapy/methods , Neoplasms/drug therapy , Active Transport, Cell Nucleus , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Products, tat/chemistry , Gene Products, tat/genetics , Gene Products, tat/metabolism , Genetic Vectors , Green Fluorescent Proteins , HeLa Cells , Humans , Kinetics , Luminescent Proteins/metabolism , Neoplasms/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use
18.
Mol Cells ; 19(1): 88-96, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15750345

ABSTRACT

Reactive oxygen species (ROS) are implicated in reperfusion injury after transient focal cerebral ischemia. The antioxidant enzyme, Cu,Zn-superoxide dismutase (SOD), is one of the major means by which cells counteract the deleterious effects of ROS after ischemia. Recently, we reported that when Tat-SOD fusion protein is transduced into pancreatic beta cells it protects the beta cells from destruction by relieving oxidative stress in ROS-implicated diabetes (Eum et al., 2004). In the present study, we investigated the protective effects of Tat-SOD fusion protein against neuronal cell death and ischemic insults. When Tat-SOD was added to the culture medium of neuronal cells, it rapidly entered the cells and protected them against paraquat-induced cell death. Immunohistochemical analysis revealed that Tat-SOD injected intraperitoneally (i.p.) into mice has access to various tissues including brain neurons. When i.p. injected into gerbils, Tat-SOD prevented neuronal cell death in the hippocampus in response to transient fore-brain ischemia. These results suggest that Tat-SOD provides a strategy for therapeutic delivery in various hu-man diseases, including stroke, related to this anti-oxidant enzyme or to ROS.


Subject(s)
Astrocytes/cytology , Astrocytes/drug effects , Ischemic Attack, Transient/drug therapy , Reperfusion Injury/prevention & control , Superoxide Dismutase/therapeutic use , Animals , Antioxidants/therapeutic use , Cell Death/drug effects , Gene Products, tat/therapeutic use , Gerbillinae , Male , Mice , Mice, Inbred ICR , Oxidative Stress/physiology , Recombinant Fusion Proteins/therapeutic use , Transduction, Genetic
19.
Hum Gene Ther ; 12(13): 1623-37, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11535166

ABSTRACT

DNA plasmid immunization has the important advantage over traditional vaccines of making it possible to combine selected genes into one vaccine. The efficacy of a combination of DNA plasmids encoding the nef, rev, and tat HIV-1 regulatory genes in inducing cellular immune responses was analyzed in asymptomatic HIV-1-infected patients. Patients initially selected for having low or no detectable immune responses to Nef, Rev, or Tat antigens developed MHC class I-restricted cytolytic activities as well as enhanced bystander effects. The induction of memory cells against target cells infected with the whole HIV-1 genome was analyzed by using a pseudovirus HIV-1/murine leukemia virus (MuLV), and target cells infected with vaccinia virus carrying the respective gene. The most remarkable change observed after immunization with the gene combination was an increase in cytotoxic T lymphocyte (CTL) precursors to target cells infected with the whole HIV-1 genome. Infection by the pseudotype HIV-1/MuLV virus should result in a multitude of HIV-1 peptides presented on the target cell surface, representative of the in vivo situation. An in vitro assessment of the expression of the single and combined gene products showed that this was consistent with the induction of CTL responses in vivo. No clinical advantage or adverse effects were noted. Therapeutic effects of such immunization may become measurable by structured therapy interruption.


Subject(s)
AIDS Vaccines/genetics , AIDS Vaccines/immunology , HIV Antigens/genetics , HIV Infections/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/therapeutic use , CD4 Lymphocyte Count , CpG Islands/genetics , Cytotoxicity, Immunologic , Gene Expression , Gene Products, nef/biosynthesis , Gene Products, nef/genetics , Gene Products, nef/immunology , Gene Products, nef/therapeutic use , Gene Products, rev/biosynthesis , Gene Products, rev/genetics , Gene Products, rev/immunology , Gene Products, rev/therapeutic use , Gene Products, tat/biosynthesis , Gene Products, tat/genetics , Gene Products, tat/immunology , Gene Products, tat/therapeutic use , Genes, Viral/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HIV Antigens/biosynthesis , HIV Antigens/immunology , HIV Infections/therapy , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , HeLa Cells , Histocompatibility Antigens Class I/immunology , Humans , Leukemia Virus, Murine/genetics , Lymphocyte Activation , Plasmids/genetics , T-Lymphocytes, Cytotoxic/cytology , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/therapeutic use , Vaccinia virus/genetics , nef Gene Products, Human Immunodeficiency Virus , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
20.
Stroke ; 34(5): 1304-10, 2003 May.
Article in English | MEDLINE | ID: mdl-12677018

ABSTRACT

BACKGROUND AND PURPOSE: Delivery of therapeutic proteins into tissues and across the blood-brain barrier is severely limited by their size and biochemical properties. The 11-amino acid human immunodeficiency virus TAT protein transduction domain is able to cross cell membranes and the blood-brain barrier, even when coupled with larger peptides. The present studies were done to evaluate whether TAT-glial line-derived neurotrophic factor (GDNF) fusion protein is protective in focal cerebral ischemia. METHODS: Anesthetized male C57BL/6j mice were submitted to intraluminal thread occlusion of the middle cerebral artery. Reperfusion was initiated 30 minutes later by thread retraction. Laser Doppler flow was monitored during the experiments. TAT-GDNF, TAT-GFP (0.6 nmol each), or vehicle was intravenously applied over 10 minutes immediately after reperfusion. After 3 days (30 minutes of ischemia), animals were reanesthetized and decapitated. Brain injury was evaluated by histochemical stainings. RESULTS: Immunocytochemical experiments confirmed the presence of TAT-GDNF protein in the brains of fusion protein-treated nonischemic control animals 3 to 4 hours after TAT fusion protein delivery. TAT-GDNF significantly reduced the number of caspase-3-immunoreactive and DNA-fragmented cells and increased the number of viable neurons in the striatum, where disseminated tissue injury was observed, compared with TAT-GFP- or vehicle-treated animals. CONCLUSIONS: Our results demonstrate that TAT fusion proteins are powerful tools for the treatment of focal ischemia when delivered both before and after an ischemic insult. This approach may be of clinical interest because such fusion proteins can be intravenously applied and reach the ischemic brain regions. This approach may therefore offer new perspectives for future strategies in stroke therapy.


Subject(s)
Brain Ischemia/drug therapy , Gene Products, tat/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Nerve Growth Factors/therapeutic use , Neuroprotective Agents/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Blood-Brain Barrier , Brain Ischemia/etiology , Brain Ischemia/pathology , Drug Administration Schedule , Drug Evaluation, Preclinical , Gene Products, tat/administration & dosage , Gene Products, tat/pharmacokinetics , Gene Products, tat/pharmacology , Genes, tat , Glial Cell Line-Derived Neurotrophic Factor , HIV-1/genetics , Infarction, Middle Cerebral Artery/complications , Infusions, Intravenous , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/pharmacokinetics , Nerve Growth Factors/pharmacology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Reperfusion Injury/pathology , Reproducibility of Results , Single-Blind Method , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL