Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Eur J Pharmacol ; 882: 173235, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32574672

ABSTRACT

Induction of Hypoxia Inducible Factor (HIF) as a direct consequence of oxygen deficiency in tumor tissues is a potent stimulus of CD73 (ecto-5'-nucleotidase) expression. Hypoxic environment and CD73 overexpression are associated with altered metabolism, elevated cancer cell proliferation, and tumor vascularization. Herein, a delivery system was developed for silencing CD73 and HIF-1α gene using siRNA-loaded Superparamagnetic iron oxide (SPION) nanocarriers for cancer treatment. SPIONs were encapsulated with thiolated chitosan (TC) and trimethyl chitosan (TMC) for improving their stabilization and functionalization. The nanoparticles (NPs) were about 133 nm in size, spherical, and non-toxic, and the addition of TAT peptide (derived from HIV-1 TAT protein) to TMC-TC-SPIONs significantly increased their cellular uptake by cancer cells. The produced NPs could efficiently accumulate in the tumor site, indicating their stability and targeting ability in reaching the tumor region. TAT-conjugated TMC-TC-SPIONs containing siRNAs could significantly reduce the HIF-1α and CD73 expression levels in cancer cells. Following transfection, cancer cells showed a significant reduction in migration and proliferation. Moreover, siRNA-loaded NPs could effectively reduce tumor growth and angiogenesis, as investigated by the chick chorioallantoic membrane (CAM) assay. This study suggested that TAT-TMC-TC-SPIONs can be potential nanocarrier for gene transfection in cancer therapy. Moreover, the co-silencing of CD73 and HIF-1α can be assumed as a novel anti-cancer treatment strategy with high tumor suppression potential.


Subject(s)
5'-Nucleotidase/genetics , Chitosan/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Magnetic Iron Oxide Nanoparticles/administration & dosage , Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , 5'-Nucleotidase/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor , Cell Survival/drug effects , Chitosan/chemistry , Chitosan/pharmacokinetics , Disease Progression , Drug Liberation , Female , Gene Expression Regulation, Neoplastic/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Magnetic Iron Oxide Nanoparticles/chemistry , Mice, Inbred BALB C , Neoplasms/genetics , Neoplasms/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacokinetics , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/pharmacokinetics
2.
Biotechnol Lett ; 42(4): 505-517, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31974645

ABSTRACT

The aim of the present study was to evaluate the efficacy of a novel DNA priming-protein boosting regimen in simultaneous enhancing humoral and cellular immunogenicity of the HIV-1-Tat-based candidate vaccines in mice. The experimental BALB/c mice were successfully immunized with the HIV-1-Tat DNA vaccine and boosted with the corresponding protein vaccine over a two-week interval and the elicitation of cellular and humoral immune responses were simultaneously assessed. The results showed that the prime-boost immunization has significantly given rise to lymphocyte proliferation and CTL responses, as well as the levels of both IgG and IgG antibodies compared to the other candidate vaccines. The results of the Th polarization also revealed that the Th1: Th2 indexes in the mice vaccinated with the HIV-1 Tat protein, Tat DNA, and the prime-boost vaccines were 1.03, 1.19, and 1.25, respectively. The results suggest that co-administration of the HIV-1-Tat DNA with the corresponding protein may serve as a potential formulation for enhancing of Tat vaccineinduced immunity and has measurable effects on shaping vaccines' induced Th polarization.


Subject(s)
Immunoglobulin G/metabolism , Lymphocytes/metabolism , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , Animals , Immunity, Cellular , Immunity, Humoral , Immunization , Immunization, Secondary , Mice , Mice, Inbred BALB C , Models, Animal , tat Gene Products, Human Immunodeficiency Virus/immunology
3.
Horm Behav ; 119: 104649, 2020 03.
Article in English | MEDLINE | ID: mdl-31821792

ABSTRACT

The majority of HIV+ patients present with neuroendocrine dysfunction and ~50% experience co-morbid neurological symptoms including motor, affective, and cognitive dysfunction, collectively termed neuroHIV. In preclinical models, the neurotoxic HIV-1 regulatory protein, trans-activator of transcription (Tat), promotes neuroHIV pathology that can be exacerbated by opioids. We and others find gonadal steroids, estradiol (E2) or progesterone (P4), to rescue Tat-mediated pathology. However, the combined effects of Tat and opioids on neuroendocrine function and the subsequent ameliorative capacity of gonadal steroids are unknown. We found that conditional HIV-1 Tat expression in naturally-cycling transgenic mice dose-dependently potentiated oxycodone-mediated psychomotor behavior. Tat increased depression-like behavior in a tail-suspension test among proestrous mice, but decreased it among diestrous mice (who already demonstrated greater depression-like behavior); oxycodone reversed these effects. Combined Tat and oxycodone produced apparent behavioral disinhibition of anxiety-like responding which was greater on diestrus than on proestrus. These mice made more central entries in an open field, but spent less time there and demonstrated greater circulating corticosterone. Tat increased the E2:P4 ratio of circulating steroids on diestrus and acute oxycodone attenuated this effect, but repeated oxycodone exacerbated it. Corticotropin-releasing factor was increased by Tat expression, acute oxycodone exposure, and was greater on diestrus compared to proestrus. In human neuroblastoma cells, Tat exerted neurotoxicity that was ameliorated by E2 (1 or 10 nM) or P4 (100, but not 10 nM) independent of oxycodone. Oxycodone decreased gene expression of estrogen and κ-opioid receptors. Thus, neuroendocrine function may be an important target for HIV-1 Tat/opioid interactions.


Subject(s)
Gonads/drug effects , Hypothalamo-Hypophyseal System/drug effects , Neurotoxicity Syndromes , Oxycodone/adverse effects , Pituitary-Adrenal System/drug effects , tat Gene Products, Human Immunodeficiency Virus/adverse effects , Animals , Anxiety/physiopathology , Anxiety/psychology , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/pathology , Cognitive Dysfunction/physiopathology , Drug Combinations , Female , Gonadal Steroid Hormones/physiology , Gonads/physiology , HIV Infections/complications , HIV Infections/physiopathology , HIV Infections/psychology , HIV-1/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Mice , Mice, Transgenic , Mood Disorders/chemically induced , Mood Disorders/pathology , Mood Disorders/physiopathology , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/physiopathology , Neurotoxicity Syndromes/psychology , Oxycodone/administration & dosage , Pituitary-Adrenal System/physiology , Psychomotor Disorders/chemically induced , Psychomotor Disorders/pathology , Psychomotor Disorders/physiopathology , Tumor Cells, Cultured , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/genetics
4.
Int J Pharm ; 565: 233-241, 2019 Jun 30.
Article in English | MEDLINE | ID: mdl-31075439

ABSTRACT

We propose a polymeric nanovehicles (PNVs)-based enhanced transdermal delivery platform. A technical advance can be found in that delivery efficiency is significantly enhanced by effective adhesion of PNVs to the cell membrane, which is characterized noninvasively by using a confocal laser scanning microscopy (CLSM)-based skin visualization technique. To this end, the PNVs with a soft core phase were fabricated through co-assembly of two amphiphilic triblock copolymers, poly(ethylene oxide)-b-poly(ε-caprolactone)-b-poly(ethylene oxide) (PEO-b-PCL-b-PEO) and poly(ethylene oxide)-b-poly(propylene oxide)-b-poly(ethylene oxide) (PEO-b-PPO-b-PEO). The softness of PNVs was tuned successfully, while maintaining the particle size at ∼110 nm, by incorporation of PEO-b-PPO-b-PEO into the PNVs to a volume fraction of 0.3. Through an ex vivo skin penetration test, we showed that transactivating transcriptional activator (TAT)-decorated soft PNVs could not only exert strong adhesion to skin but also increase cellular uptake, leading to a transdermal delivery efficiency that is twice that of a hard PNV control. Moreover, CLSM-based noninvasive visualization of a fluorescent drug probe in the skin showed that the adhesiveness and softness of the PNVs contributed directly to the enhancement of transdermal delivery.


Subject(s)
Cell Membrane , Drug Delivery Systems , Peptide Fragments/administration & dosage , Polyesters/administration & dosage , Polyethylene Glycols/administration & dosage , Propylene Glycols/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , Adhesiveness , Administration, Cutaneous , Animals , Cell Line, Tumor , Humans , Microscopy, Confocal , Particle Size , Polyesters/chemistry , Polyethylene Glycols/chemistry , Propylene Glycols/chemistry , Skin/metabolism , Skin Absorption , Swine
5.
Mar Drugs ; 17(5)2019 May 12.
Article in English | MEDLINE | ID: mdl-31083641

ABSTRACT

As the first in a new class of non-opioid drugs, ω-Conotoxin MVIIA was approved for the management of severe chronic pains in patients who are unresponsive to opioid therapy. Unfortunately, clinical application of MVIIA is severely limited due to its poor ability to penetrate the blood-brain barrier (BBB), reaching the central nervous system (CNS). In the present study, we have attempted to increase MVIIA's ability to cross the BBB via a fusion protein strategy. Our results showed that when the TAT-transducing domain was fused to the MVIIA C-terminal with a linker of varied numbers of glycine, the MVIIA-TAT fusion peptide exhibited remarkable ability to cross the bio-membranes. Most importantly, both intravenous and intranasal administrations of MVIIA-TAT in vivo showed therapeutic efficacy of analgesia. Compared to the analgesic effects of intracerebral administration of the nascent MVIIA, these systemic administrations of MVIIA-TAT require higher doses, but have much prolonged effects. Taken together, our results showed that TAT conjugation of MVIIA not only enables its peripheral administration, but also maintains its analgesic efficiency with a prolonged effective time window. Intranasal administration also rendered the MVIIA-TAT advantages of easy applications with potentially reduced side effects. Our results may present an alternative strategy to improve the CNS accessibility for neural active peptides.


Subject(s)
Analgesics/pharmacokinetics , Blood-Brain Barrier/metabolism , Calcium Channel Blockers/pharmacology , Recombinant Fusion Proteins/pharmacokinetics , omega-Conotoxins/pharmacokinetics , tat Gene Products, Human Immunodeficiency Virus/pharmacokinetics , Analgesics/administration & dosage , Animals , Blood-Brain Barrier/drug effects , Female , Male , Mice , Pain/drug therapy , Pain/metabolism , Pain Measurement/drug effects , Peptides/administration & dosage , Peptides/chemistry , RNA-Binding Protein FUS , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/biosynthesis , Tremor/drug therapy , Tremor/metabolism , omega-Conotoxins/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
6.
Brain Behav Immun ; 80: 227-237, 2019 08.
Article in English | MEDLINE | ID: mdl-30872089

ABSTRACT

While the advent of combination antiretroviral therapy (cART) has dramatically increased the lifespan of people living with HIV-1 paradoxically, the prevalence of NeuroHIV in people treated with cART is on the rise. It has been well documented that despite the effectiveness of cART in suppressing viremia, CNS continues to harbor viral reservoirs with persistent low-level virus replication. This, in turn, leads to the presence and accumulation of early viral protein - HIV-1 Tat, that is a well-established cytotoxic agent. In the current study, we demonstrated that exposure of mouse microglia to HIV-1 Tat resulted both in a dose- and time-dependent upregulation of miRNA-34a, with concomitant downregulation of NLRC5 (a negative regulator of NFκB signaling) expression. Using bioinformatics analyses and Argonaute immunoprecipitation assay NLRC5 was identified as a novel target of miRNA-34a. Transfection of mouse primary microglia with miRNA-34a mimic significantly downregulated NLRC5 expression, resulting in increased expression of NFκB p65. In contrast, transfection of cells with miRNA-34a inhibitor upregulated NLRC5 levels. Using pharmacological approaches, our findings showed that HIV-1 Tat-mediated microglial activation involved miRNA-34a-mediated downregulation of NLRC5 with concomitant activation of NFκB signaling. Reciprocally, inhibition of miRNA-34a blocked HIV-1 Tat-mediated microglial activation. In summary, our findings identify yet another novel mechanism of HIV-1 Tat-mediated activation of microglia involving the miRNA-34a-NLRC5-NFκB axis. These in vitro findings were also validated in the medial prefrontal cortices of HIV-1 transgenic rats as well as in SIV-infected rhesus macaques. Overall, these findings reveal the involvement of miRNA-34a-NLRC5-NFκB signaling axis in HIV-1 Tat-mediated microglial inflammation.


Subject(s)
Encephalitis/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Microglia/metabolism , NF-kappa B/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Female , Macaca mulatta , Male , Prefrontal Cortex/metabolism , Primary Cell Culture , Rats, Sprague-Dawley , Rats, Transgenic , Signal Transduction , Up-Regulation , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
7.
J Neuroimmune Pharmacol ; 14(3): 375-382, 2019 09.
Article in English | MEDLINE | ID: mdl-30905008

ABSTRACT

HIV-1 infection causes chronic neuroinflammation resulting in cognitive decline associated with diminution of survival of neural stem cells (NSC). In part, this is attributable to production of toxic viral proteins (gp120 and tat) by infected cells in the brain that can activate microglia. Here, we evaluated a novel model for HIV-1 neuropathogenesis by direct administration of viral proteins into the hippocampus. Chronic administration of either HIV-1 gp120 or tat over 14 days significantly decreased NSC proliferation, survival and neuroblast formation (by 32-37%) within the hippocampal subgranular zone as detected by doublecortin/BrdU or Ki67-positive cells. Intrahippocampal administration of gp120 or tat induced microglial activation within the hippocampus as determined by increases in microglial number and increases in the volume of the microglia (2.5-3-fold, evaluated by double IBA-1/CD68 staining). We further assessed inflammatory responses within the hippocampus by RNAseq and Ingenuity Pathway Analysis. There was a significant mRNA upregulation of numerous inflammatory mediators including Il1b, Icam1, Il12a, Ccl2, and Ccl4. These data suggest that chronic administration induces a prolonged inflammatory state within the hippocampus that negatively affects NSC survival potentially leading to cognitive dysfunction. Graphical Abstract.


Subject(s)
AIDS Dementia Complex/etiology , Disease Models, Animal , HIV Envelope Protein gp120/toxicity , HIV-1/pathogenicity , Hippocampus/drug effects , Inflammation/chemically induced , Neural Stem Cells/drug effects , tat Gene Products, Human Immunodeficiency Virus/toxicity , Animals , HIV Envelope Protein gp120/administration & dosage , Hippocampus/metabolism , Hippocampus/pathology , Inflammation Mediators/metabolism , Infusions, Parenteral , Mice , Mice, Inbred C57BL , Microglia/physiology , Neural Stem Cells/pathology , Neurogenesis , Random Allocation , Recombinant Proteins/administration & dosage , Recombinant Proteins/toxicity , Sequence Analysis, RNA , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
8.
Biomacromolecules ; 19(7): 2682-2690, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29847726

ABSTRACT

We herein propose a polymeric nanovehicle system that has the ability to remarkably improve cellular uptake and transdermal delivery. Cell-penetrating peptide-patchy deformable polymeric nanovehicles were fabricated by tailored coassembly of amphiphilic poly(ethylene oxide)- block-poly(ε-caprolactone) (PEO- b-PCL), mannosylerythritol lipid (MEL), and YGRKKRRQRRR-cysteamine (TAT)-linked MEL. Using X-ray diffraction, differential scanning calorimetry, and nuclear magnetic resonance analyses, we revealed that the incorporation of MEL having an asymmetric alkyl chain configuration was responsible for the deformable phase property of the vehicles. We also discovered that the nanovehicles were mutually attracted, exhibiting a gel-like fluid characteristic due to the dipole-dipole interaction between the hydroxyl group of MEL and the methoxy group of PEO- b-PCL. Coassembly of TAT-linked MEL with the deformable nanovehicles significantly enhanced cellular uptake due to macropinocytosis and caveolae-/lipid raft-mediated endocytosis. Furthermore, the in vivo skin penetration test revealed that our TAT-patchy deformable nanovehicles remarkably improved transdermal delivery efficiency.


Subject(s)
Glycolipids/chemistry , Nanoparticles/chemistry , Peptide Fragments/administration & dosage , Polyesters/chemistry , Skin Absorption , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , Administration, Cutaneous , Adult , Cell Line , Cysteamine/chemistry , Female , Humans , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/pharmacokinetics
9.
JCI Insight ; 3(7)2018 04 05.
Article in English | MEDLINE | ID: mdl-29618654

ABSTRACT

Replication competent HIV-1 persists in a subpopulation of CD4+ T lymphocytes despite prolonged antiretroviral treatment. This residual reservoir of infected cells harbors transcriptionally silent provirus capable of reigniting productive infection upon discontinuation of antiretroviral therapy. Certain classes of drugs can activate latent virus but not at levels that lead to reductions in HIV-1 reservoir size in vivo. Here, we show the utility of CD4+ receptor targeting exosomes as an HIV-1 latency reversal agent (LRA). We engineered human cellular exosomes to express HIV-1 Tat, a protein that is a potent transactivator of viral transcription. Preparations of exosomal Tat-activated HIV-1 in primary, resting CD4+ T lymphocytes isolated from antiretroviral-treated individuals with prolonged periods of viral suppression and led to the production of replication competent HIV-1. Furthermore, exosomal Tat increased the potency of selected LRA by over 30-fold in terms of HIV-1 mRNA expression, thereby establishing it as a potentially new class of biologic product with possible combinatorial utility in targeting latent HIV-1.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Drug Carriers , HIV Infections/drug therapy , Recombinant Fusion Proteins/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , Adult , Aged , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active/methods , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cell Engineering/methods , Cloning, Molecular , Exosomes , Female , HEK293 Cells , HIV Infections/blood , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/pathogenicity , Humans , Male , Middle Aged , Primary Cell Culture , Protein Engineering/methods , Proto-Oncogene Proteins c-myc/administration & dosage , Proto-Oncogene Proteins c-myc/genetics , Recombinant Fusion Proteins/genetics , Transfection , Virus Latency/drug effects , Virus Latency/immunology , Virus Replication/drug effects , Virus Replication/immunology , tat Gene Products, Human Immunodeficiency Virus/genetics
10.
J Control Release ; 261: 174-186, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28662902

ABSTRACT

Cell-penetrating peptides (CPPs) are increasingly important in transporting macromolecules across cell membranes, but their use remains confined to narrow clinical applications due to the systemic toxicity induced by their positive charges. Several newly discovered electronic neutral penetrating peptides are not attracting much attention because their penetrating capacity is normally far less powerful than cationic or amphiphilic CPPs. In this study, we found the electronic neutral cyclic peptide cyclosporin A (CsA) exhibited 5.6-fold and 19.1-fold stronger penetrating capacity, respectively, than two reported electronic neutral peptides PFVYLI (PFV) and pentapeptide VPTLQ (VPT) in MCF-7 human breast cancer cells. To systematically evaluate the efficiency and toxicity of CsA, we utilized CsA to deliver a membrane-impenetrable pro-apoptotic peptide (PAD) and compared this to the well-established cationic penetrating peptide TAT (RKKRRQRRR). By conjugating CsA to PAD, the internalization of PAD increased 2.2- to 4.7-fold in four different tumor cell lines, and that of CsA-PAD conjugate was significantly higher than TAT-PAD conjugate in MCF-7 and HeLa human cervical cancer cells. Cytotoxicity studies demonstrated that CsA-PAD exhibited a large increase in cell cytotoxicity compared to PAD in four different tumor cell lines, with the effect being similar or greater than the effect of TAT-PAD, depending upon the cell type. The mechanistic studies demonstrated that modifying CsA or TAT did not change the cytotoxicity mechanism of PAD, which occurred via mitochondrial membrane damage related to apoptosis. In vivo studies showed that CsA-PAD could achieve similar anti-tumor efficacy to TAT-PAD but with much lower systemic toxicity, especially to the heart and liver. In conclusion, our study demonstrates for the first time that the electronic-neutral penetrating peptide CsA can be used as a powerful tool to deliver peptide drugs with similar efficiency and less toxicity than the positively charged TAT peptide.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Cyclosporine/administration & dosage , Neoplasms/drug therapy , Peptides/administration & dosage , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell-Penetrating Peptides/toxicity , Cyclosporine/toxicity , Drug Delivery Systems , Female , HeLa Cells , Humans , MCF-7 Cells , Mice , Mice, Nude , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/pathology , Neoplasms/metabolism , Oligopeptides/administration & dosage , Oligopeptides/toxicity , Peptide Fragments/administration & dosage , Peptide Fragments/toxicity , Peptides/pharmacokinetics , Peptides/pharmacology , Xenograft Model Antitumor Assays , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/toxicity
11.
J Neurosci ; 37(33): 7837-7847, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28716964

ABSTRACT

HIV-associated neurocognitive disorder (HAND) affects approximately half of HIV-infected patients. Loss of synaptic connections is a hallmark of many neurocognitive disorders, including HAND. The HIV-1 protein transactivator of transcription (Tat) disrupts synaptic connections both in vitro and in vivo and has been linked to impaired neurocognitive function in humans. In vitro studies have shown that ifenprodil, an antagonist selective for GluN2B-containing NMDARs, reverses synapse loss when applied after Tat. Here, we tested the hypothesis that Tat-induced loss and ifenprodil-mediated rescue of synaptic spines in vivo would predict impairment and rescue of cognitive function. Using intracranial multiphoton imaging, we found that infusion of 100 ng of HIV-1 Tat into the lateral ventricle of yellow fluorescent protein-expressing transgenic mice produced a 17 ± 1% loss of dendritic spines in layer 1 of retrosplenial cortex. Repeated imaging of the same dendrites over 3 weeks enabled longitudinal experiments that demonstrated sustained spine loss after Tat infusion and transient rescue after ifenprodil administration (10 mg/kg, i.p.). Parallel trace fear conditioning experiments showed that spine loss predicted learning deficits and that the time course of ifenprodil-induced rescue of spine density correlated with restoration of cognitive function. These results show for the first time that, during exposure to an HIV-1 neurotoxin in vivo, alteration of GluN2B-containing NMDAR signaling suppresses spine density and impairs learning. Pharmacological inhibition of these NMDARs rescued spines and restored cognitive function. Drugs that rescue synapses may improve neurocognitive function in HAND.SIGNIFICANCE STATEMENT Synaptodendritic damage correlates with cognitive decline in HIV-associated neurocognitive disorder (HAND) patients. We developed an in vivo imaging approach for longitudinal tracking of spine density that enabled correlation of synaptic changes with behavioral outcomes in a model of HAND. We show for the first time that spine loss after exposure to an HIV-1 protein can be reversed pharmacologically and that loss and recovery of dendritic spines predict impairment and restoration of cognitive function, respectively. Therefore, synapse loss, the hallmark of cognitive decline in HAND, is reversible. Drugs that restore spine density may have broad application for improving cognitive function during the early phases of neurodegenerative diseases.


Subject(s)
Cognitive Dysfunction/prevention & control , Excitatory Amino Acid Antagonists/administration & dosage , HIV-1 , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synapses/drug effects , tat Gene Products, Human Immunodeficiency Virus/toxicity , Animals , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Infusions, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Piperidines/administration & dosage , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
12.
Oncogene ; 36(18): 2609-2618, 2017 05 04.
Article in English | MEDLINE | ID: mdl-27819680

ABSTRACT

Syndecan-1 is a heparan sulfate proteoglycan (HSPG) commonly upregulated in AIDS-related B lymphoid malignancies. Tat is the main HIV-1 transactivating factor that has a major role in the pathogenesis of AIDS-related lymphomas (ARL) by engaging heparan sulfate proteoglycans (HSPGs), chemokine receptors and integrins at the lymphoid cell (LC) surface. Here B-lymphoid Namalwa cell clones that do not express or overexpress syndecan-1 (EV-Ncs and SYN-Ncs, respectively) were compared for their responsiveness with Tat: in the absence of syndecan-1, Tat induces a limited EV-Nc migration via C-X-C motif chemokine receptor 4 (CXCR4), G-proteins and Rac. Syndecan-1 overexpression increases SYN-Nc responsiveness to Tat and makes this response independent from CXCR4 and G-protein and dependent instead on pp60src phosphorylation. Tat-induced SYN-Nc migration and pp60src phosphorylation require the engagement of αvß3 integrin and consequent pp125FAK phosphorylation. This complex set of Tat-driven activations is orchestrated by the direct interaction of syndecan-1 with pp60src and its simultaneous coupling with αvß3. The Tat/syndecan-1/αvß3 interplay is retained in vivo and is shared also by other syndecan-1+ B-LCs, including BJAB cells, whose responsiveness to Tat is inhibited by syndecan-1 knockdown. In conclusion, overexpression of syndecan-1 confers to B-LCs an increased capacity to migrate in response to Tat, owing to a switch from a CXCR4/G-protein/Rac to a syndecan-1/αvß3/pp60src/pp125FAK signal transduction pathway that depends on the formation of a complex in which syndecan-1 interacts with Tat via its HS-chains, with αvß3 via its core protein ectodomain and with pp60src via its intracellular tail. These findings have implications in ARL progression and may help in identifying new therapeutical targets for the treatment of AIDS-associated neoplasia.


Subject(s)
Focal Adhesion Kinase 1/genetics , Integrin alphaVbeta3/genetics , Lymphocytes/metabolism , Neoplasms/genetics , Syndecan-1/genetics , Cell Adhesion/genetics , Gene Expression Regulation, Neoplastic , HIV-1/genetics , Humans , Lymphocytes/pathology , Multiprotein Complexes/genetics , Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/genetics , Receptors, CXCR4/genetics , Signal Transduction/genetics , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/metabolism
13.
Sci Rep ; 6: 29131, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27358023

ABSTRACT

DNA vaccines are cost-effective to manufacture on a global scale and Tat-based DNA vaccines have yielded protective outcomes in preclinical and clinical models of human immunodeficiency virus (HIV), highlighting the potential of such vaccines. However, Tat-based DNA vaccines have been poorly immunogenic, and despite the administration of multiple doses and/or the addition of adjuvants, these vaccines are not in general use. In this study, we improved Tat immunogenicity by fusing it with the oligomerisation domain of a chimeric C4-binding protein (C4b-p), termed IMX313, resulting in Tat heptamerisation and linked Tat to the leader sequence of tissue plasminogen activator (TPA) to ensure that the bulk of heptamerised Tat is secreted. Mice vaccinated with secreted Tat fused to IMX313 (pVAX-sTat-IMX313) developed higher titres of Tat-specific serum IgG, mucosal sIgA and cell-mediated immune (CMI) responses, and showed superior control of EcoHIV infection, a surrogate murine HIV challenge model, compared with animals vaccinated with other test vaccines. Given the crucial contribution of Tat to HIV-1 pathogenesis and the precedent of Tat-based DNA vaccines in conferring some level of protection in animal models, we believe that the virologic control demonstrated with this novel multimerised Tat vaccine highlights the promise of this vaccine candidate for humans.


Subject(s)
Apoptosis Regulatory Proteins , HIV Infections , Recombinant Fusion Proteins , Vaccines, DNA , tat Gene Products, Human Immunodeficiency Virus , Animals , Humans , Mice , Apoptosis Regulatory Proteins/administration & dosage , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/drug effects , HIV-1/immunology , HIV-1/pathogenicity , Immunity, Cellular , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Survivin , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology
14.
Int J Pharm ; 511(1): 236-244, 2016 Sep 10.
Article in English | MEDLINE | ID: mdl-27363937

ABSTRACT

We have investigated the co-addition of hexadecylphosphocholine (HePC) and a Tat derived peptide (Tat), coupled to Maleimide-PEG2000-DSPE pegylated liposomal doxorubicin (PLD) in many respects, including drug and liposome cellular delivery, drug release, biodistribution, in vivo cell delivery and antitumor activity. The liposomes were HePC-free and -containing liposomes, from which liposomes with 25, 50, 100 and 200 numbers of Tat/liposome were prepared. Similarly, DiI-C18 (3)-model liposomes (DiI-L and DiI-HePC-L) were prepared. HePC and Tat increased cellular delivery of Dox and cytotoxicity in B16F0 melanoma and C26 colon carcinoma cells. Tat enhanced liposome-cell interaction and caused Dox burst release. HePC and Tat reduced the serum retention time of liposomal Dox, slightly and dramatically, respectively. In comparison, Tat-liposomes enhanced Dox delivery to liver and spleen cells 3h post-injection. Likewise, Dox content of these tissues and tumor was lower at 24h. The naïve liposomes retarded tumor growth more effectively and their related median survival time of the treated C26 bearing BALB/c mice was longer than those of Tat-liposomes (MST>45days versus MST<38days). Overall liposomes exhibiting sustained drug release and negligible cell interaction were more suitable delivery systems in targeting cancerous tumors and suppressing their growth.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/analogs & derivatives , Drug Delivery Systems/methods , Peptide Fragments/administration & dosage , Phosphorylcholine/analogs & derivatives , Polyethylene Glycols/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , Animals , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Doxorubicin/administration & dosage , Doxorubicin/metabolism , Drug Liberation , Female , Liposomes , Mice , Mice, Inbred BALB C , Peptide Fragments/metabolism , Phosphorylcholine/administration & dosage , Phosphorylcholine/metabolism , Polyethylene Glycols/metabolism , Tissue Distribution/drug effects , Tissue Distribution/physiology , Xenograft Model Antitumor Assays/methods , tat Gene Products, Human Immunodeficiency Virus/metabolism
15.
ACS Appl Mater Interfaces ; 8(28): 17944-54, 2016 Jul 20.
Article in English | MEDLINE | ID: mdl-27353012

ABSTRACT

Reactive oxygen species (ROS) are important factors in many clinical diseases. However, direct delivery of antioxidant enzymes into cells is difficult due to poor cell uptake. A proper design of delivery of enzymes by nanoparticles is very desirable for therapeutic purposes. To overcome the cell barrier problem, a designed mesoporous silica nanoparticle (MSN) system with attached TAT-fusion denatured enzyme for enhancing cell membrane penetration has been developed. Simultaneous delivery of two up-downstream antioxidant enzymes, superoxide dismutase (SOD) and glutathione peroxidase(GPx), reveals synergistic efficiency of ROS scavenging, compared to single antioxidant enzyme delivery. TAT peptide conjugation provided a facile nonendocytosis cell uptake and escape from endosome while moving and aggregating along the cytoskeleton that would allow them to be close to each other at the same time, resulting in the cellular antioxidation cascade reaction. The two-enzyme delivery shows a significant synergistic effect for protecting cells against ROS-induced cell damage and cell cycle arrest. The nanocarrier strategy for enzyme delivery demonstrates that intracellular anti-ROS cascade reactions could be regulated by multifunctional MSNs carrying image fluorophore and relevant antioxidation enzymes.


Subject(s)
Antioxidants/administration & dosage , Glutathione Peroxidase/administration & dosage , Nanoparticles/chemistry , Recombinant Fusion Proteins/administration & dosage , Superoxide Dismutase/administration & dosage , Antioxidants/chemistry , Glutathione Peroxidase/chemistry , HeLa Cells , Humans , Nanoparticles/administration & dosage , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Protein Denaturation , Recombinant Fusion Proteins/chemistry , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Superoxide Dismutase/chemistry , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/chemistry
16.
Int J Pharm ; 501(1-2): 112-23, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-26828670

ABSTRACT

The dual-ligand dendritic polyamidoamine-(polyethylene glycol)n-cyclic RGDyC peptide-(TAT peptide) (PPnR(T)) with various supplied molar ratios of polyethylene glycol (PEG) to polyamidoamine (PAMAM) (n=5, 15, 30) were designed as drug-carriers for the treatment of neovascular diseases; their targeting and penetrating effects were subsequently evaluated. (1)H NMR demonstrated PPnR(T) was successfully synthesized. Compared with the unmodified PAMAM, in vitro cytotoxicity of PPnR(T) to αvß3 negative cells (αvß3-) was significantly reduced, whereas the lethality to pathologic neovascular endothelial cells (αvß3+) was efficiently increased compared to PPn. Compared to PP5R(T) and PP15R(T), PP30R(T) exhibited the most selective and efficient cellular uptake by human umbilical vein endothelial cells (HUVECs, αvß3+). Membrane interaction study indicated the cellular uptake process of PP30R(T) of HUVECs mainly involved specific RGD-αvß3 recognition as well as electrostatic interactions. Intracellular localization results confirmed PP30R(T) was distributed in the cytoplasm in HUVECs. 3D tumor spheroids penetration studies demonstrated that PP30R(T) penetrated the A549 cells to reach the depths of the avascular tumor spheroids. In vivo imaging further demonstrated that PP30R(T) achieved profoundly improved distribution in tumor tissues where angiogenesis existed. Therefore, the bi-functional dendrimer PP30R(T) displayed great potential as a nano-carrier for targeted drug delivery both in vitro and in vivo, and had broad prospects as nanocarriers for the targeted treatment of neovascular diseases.


Subject(s)
Cell-Penetrating Peptides , Dendrimers , Oligopeptides , Peptide Fragments , Peptides, Cyclic , tat Gene Products, Human Immunodeficiency Virus , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Cells, Cultured , Dendrimers/administration & dosage , Dendrimers/chemistry , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Maleimides/chemistry , Mice, Inbred BALB C , Mice, Nude , Nanostructures/administration & dosage , Nanostructures/chemistry , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemistry , Polyethylene Glycols/chemistry , Succinimides/chemistry , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/chemistry
17.
Int J Nanomedicine ; 11: 501-12, 2016.
Article in English | MEDLINE | ID: mdl-26893558

ABSTRACT

Gene therapy has promising applications in ovarian cancer therapy. Blocking the function of the survivin protein could lead to the growth inhibition of cancer cells. Herein, we used degradable heparin-polyethyleneimine (HPEI) nanoparticles to deliver a dominant-negative human survivin T34A (hs-T34A) gene to treat ovarian cancer. HPEI nanoparticles were characterized and were found to have a dynamic diameter of 66±4.5 nm and a zeta potential of 27.1±1.87 mV. The constructed hs-T34A gene expression plasmid could be effectively delivered into SKOV3 ovarian carcinoma cells by HPEI nanoparticles with low cytotoxicity. Intraperitoneal administration of HPEI/hs-T34A complexes could markedly inhibit tumor growth in a mouse xenograft model of SKOV3 human ovarian cancer. Moreover, according to our results, apparent apoptosis of cancer cells was observed both in vitro and in vivo. Taken together, the prepared HPEI/hs-T34A formulation showed potential applications in ovarian cancer gene therapy.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Genetic Therapy/methods , Nanoparticles , Ovarian Neoplasms/therapy , Recombinant Fusion Proteins/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/administration & dosage , Female , Heparin/chemistry , Humans , Injections, Intraperitoneal , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Plasmids/genetics , Polyethyleneimine/chemistry , Rats, Sprague-Dawley , Recombinant Fusion Proteins/administration & dosage , Survivin , Tissue Distribution , Xenograft Model Antitumor Assays , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
18.
Nano Lett ; 16(2): 1509-13, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26745653

ABSTRACT

Semiconductor nanowire (NW) devices that can address intracellular electrophysiological events with high sensitivity and spatial resolution are emerging as key tools in nanobioelectronics. Intracellular delivery of NWs without compromising cellular integrity and metabolic activity has, however, proven difficult without external mechanical forces or electrical pulses. Here, we introduce a biomimetic approach in which a cell penetrating peptide, the trans-activating transcriptional activator (TAT) from human immunodeficiency virus 1, is linked to the surface of Si NWs to facilitate spontaneous internalization of NWs into primary neuronal cells. Confocal microscopy imaging studies at fixed time points demonstrate that TAT-conjugated NWs (TAT-NWs) are fully internalized into mouse hippocampal neurons, and quantitative image analyses reveal an ca. 15% internalization efficiency. In addition, live cell dynamic imaging of NW internalization shows that NW penetration begins within 10-20 min after binding to the membrane and that NWs become fully internalized within 30-40 min. The generality of cell penetrating peptide modification method is further demonstrated by internalization of TAT-NWs into primary dorsal root ganglion (DRG) neurons.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Drug Delivery Systems , Nanowires/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , Animals , Cell-Penetrating Peptides/chemistry , Humans , Mice , Microscopy, Confocal , Nanowires/chemistry , Neurons/drug effects , Neurons/ultrastructure , Primary Cell Culture , Semiconductors , tat Gene Products, Human Immunodeficiency Virus/chemistry
19.
Mol Biotechnol ; 58(1): 22-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26646387

ABSTRACT

Neuroglobin (NGB) is a newly discovered neuroprotector and mainly localized in the neurons and retinal cells of the central and peripheral nervous systems in vertebrates, and its prokaryotic expression protein of which fused with HIV-1 virus-encoded Tat peptide exhibited significant antioxidant and anti-hypoxia. However, no study has documented on the anti-hypoxia of yeast expressed Tat-NGB. To address it, the NGB cDNA fragment with and without Tat tag was designed and conjugated to pPIC9K followed by electroporation, and positive colonies were screened. Subsequently, Tat-NGB-His and His-NGB-His proteins were expressed by inducer methanol and identified by SDS-PAGE, and purified with HisTrap™ FF crude column. After desalting, the transmembrane transduction of Tat-NGB was examined and identified by Western blot, and the anti-hypoxia activity was also examined by CCK-8 kit. Unexpectedly, Tat-NGB-His and His-NGB-His proteins were high yield and secretory expressed in GS115 Pichia pastoris. After purification, the high purified protein was prepared and exhibited a significant transmembrane transduction of Tat-NGB-His (**p < 0.01, compare to control and His-NGB-His). Significantly, Tat-NGB-His could protect hypoxia induced injury of PC12 cells and had an obviously difference when comparing to control and His-NGB-His groups (*p < 0.05, **p < 0.01). The present study first reported the yeast expressed production of Tat-NGB-His and His-NGB-His, and then elucidated the transduction and neuroprotection of Tat-NGB-His on PC12 cell. It not only provided a significant reference for high-yield expression of NGB in yeast expression system, but also provided a significant prevention and treatment of hypoxic and ischemic brain injury.


Subject(s)
Globins/biosynthesis , Hypoxia-Ischemia, Brain/genetics , Nerve Tissue Proteins/biosynthesis , Recombinant Fusion Proteins/biosynthesis , tat Gene Products, Human Immunodeficiency Virus/biosynthesis , Animals , Cell Hypoxia/drug effects , Gene Expression Regulation/drug effects , Globins/administration & dosage , Globins/genetics , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/pathology , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/genetics , Neuroglobin , Neuroprotective Agents/administration & dosage , PC12 Cells , Pichia , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Transduction, Genetic , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/genetics
20.
J Drug Target ; 24(1): 13-23, 2016.
Article in English | MEDLINE | ID: mdl-26061295

ABSTRACT

Therapeutics targeting the Nogo-A signal pathway hold promise to promote recovery following brain injury. Based on the temporal characteristics of Nogo-A expression in the process of cerebral ischemia and reperfusion, we tested a novel asynchronous treatment, in which TAT-M9 was used in the early stage to decrease neuronal loss, and TAT-NEP1-40 was used in the delayed stage to promote neurite outgrowth after bilateral common carotid artery occlusion (BCCAO) in mice. Both TAT-M9 and TAT-NEP1-40 were efficiently delivered into the brains of mice by intraperitoneal injection. TAT-M9 treatment promoted neuron survival and inhibited neuronal apoptosis. Asynchronous therapy with TAT-M9 and TAT-NEP1-40 increased the expression of Tau, GAP43 and MAP-2 proteins, and enhanced short-term and long-term cognitive functions. In conclusion, the asynchronous treatment had a long-term neuroprotective effect, which reduced neurologic injury and apoptosis, promoted neurite outgrowth and enhanced functional recovery after ischemia. It suggests that this asynchronous treatment could be a promising therapy for cerebral ischemia in humans.


Subject(s)
Brain Ischemia/physiopathology , Myelin Proteins/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , tat Gene Products, Human Immunodeficiency Virus/pharmacology , Animals , Apoptosis/drug effects , Behavior Rating Scale , Cell Survival/drug effects , Disease Models, Animal , Drug Administration Schedule , GAP-43 Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Myelin Proteins/administration & dosage , Myelin Proteins/pharmacology , Neurites/drug effects , Nogo Proteins , Peptide Fragments/administration & dosage , Random Allocation , Reperfusion Injury/physiopathology , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...