Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.128
1.
Int J Mol Sci ; 25(6)2024 Mar 16.
Article En | MEDLINE | ID: mdl-38542351

Viruses provide vital insights into gene expression control. Viral transactivators, with other viral and cellular proteins, regulate expression of self, other viruses, and host genes with profound effects on infected cells, underlying inflammation, control of immune responses, and pathogenesis. The multifunctional Tat proteins of lentiviruses (HIV-1, HIV-2, and SIV) transactivate gene expression by recruiting host proteins and binding to transacting responsive regions (TARs) in viral and host RNAs. SARS-CoV-2 nucleocapsid participates in early viral transcription, recruits similar cellular proteins, and shares intracellular, surface, and extracellular distribution with Tat. SARS-CoV-2 nucleocapsid interacting with the replication-transcription complex might, therefore, transactivate viral and cellular RNAs in the transcription and reactivation of self and other viruses, acute and chronic pathogenesis, immune evasion, and viral evolution. Here, we show, by using primary and secondary structural comparisons, that the leaders of SARS-CoV-2 and other coronaviruses contain TAR-like sequences in stem-loops 2 and 3. The coronaviral nucleocapsid C-terminal domains harbor a region of similarity to TAR-binding regions of lentiviral Tat proteins, and coronaviral nonstructural protein 12 has a cysteine-rich metal binding, dimerization domain, as do lentiviral Tat proteins. Although SARS-CoV-1 nucleocapsid transactivated gene expression in a replicon-based study, further experimental evidence for coronaviral transactivation and its possible implications is warranted.


COVID-19 , HIV-1 , Humans , HIV-1/physiology , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Transcriptional Activation , HIV Long Terminal Repeat , COVID-19/genetics , Gene Products, tat/genetics , Lentivirus/genetics , Gene Expression , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , RNA, Viral/metabolism
2.
Nat Commun ; 14(1): 8397, 2023 Dec 18.
Article En | MEDLINE | ID: mdl-38110433

The development of latency reversing agents that potently reactivate HIV without inducing global T cell activation would benefit the field of HIV reservoir research and could pave the way to a functional cure. Here, we explore the reactivation capacity of a lipid nanoparticle containing Tat mRNA (Tat-LNP) in CD4 T cells from people living with HIV undergoing antiretroviral therapy (ART). When combined with panobinostat, Tat-LNP induces latency reversal in a significantly higher proportion of latently infected cells compared to PMA/ionomycin (≈ 4-fold higher). We demonstrate that Tat-LNP does not alter the transcriptome of CD4 T cells, enabling the characterization of latently infected cells in their near-native state. Upon latency reversal, we identify transcriptomic differences between infected cells carrying an inducible provirus and non-infected cells (e.g. LINC02964, GZMA, CCL5). We confirm the transcriptomic differences at the protein level and provide evidence that the long non-coding RNA LINC02964 plays a role in active HIV infection. Furthermore, p24+ cells exhibit heightened PI3K/Akt signaling, along with downregulation of protein translation, suggesting that HIV-infected cells display distinct signatures facilitating their long-term persistence. Tat-LNP represents a valuable research tool for in vitro reservoir studies as it greatly facilitates the in-depth characterization of HIV reservoir cells' transcriptome and proteome profiles.


Gene Products, tat , HIV-1 , Nanoparticles , RNA, Viral , Virus Latency , Virus Latency/drug effects , Virus Latency/genetics , Gene Products, tat/genetics , Gene Products, tat/metabolism , RNA, Viral/administration & dosage , RNA, Viral/genetics , RNA, Viral/metabolism , Nanoparticles/administration & dosage , Nanoparticles/chemistry , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/virology , Panobinostat/pharmacology , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , CD4 Antigens/genetics , CD4 Antigens/metabolism , HIV-1/drug effects , HIV-1/genetics , Proviruses/drug effects , Proviruses/genetics , Single-Cell Gene Expression Analysis , HIV Core Protein p24/genetics , HIV Core Protein p24/metabolism , RNA, Long Noncoding/metabolism , Cells, Cultured , Humans , Ionomycin/pharmacology
3.
Neurochem Int ; 167: 105538, 2023 Jul.
Article En | MEDLINE | ID: mdl-37207854

Oxidative stress plays a key role in the pathogenesis of neuronal injury, including ischemia. Ras-related nuclear protein (RAN), a member of the Ras superfamily, involves in a variety of biological roles, such as cell division, proliferation, and signal transduction. Although RAN reveals antioxidant effect, its precise neuroprotective mechanisms are still unclear. Therefore, we investigated the effects of RAN on HT-22 cell which were exposed to H2O2-induced oxidative stress and ischemia animal model by using the cell permeable Tat-RAN fusion protein. We showed that Tat-RAN transduced into HT-22 cells, and markedly inhibited cell death, DNA fragmentation, and reactive oxygen species (ROS) generation under oxidative stress. This fusion protein also controlled cellular signaling pathways, including mitogen-activated protein kinases (MAPKs), NF-κB, and apoptosis (Caspase-3, p53, Bax and Bcl-2). In the cerebral forebrain ischemia animal model, Tat-RAN significantly inhibited both neuronal cell death, and astrocyte and microglia activation. These results indicate that RAN significantly protects against hippocampal neuronal cell death, suggesting Tat-RAN will help to develop the therapies for neuronal brain diseases including ischemic injury.


Brain Injuries , Brain Ischemia , Neuroprotective Agents , Animals , Hydrogen Peroxide/pharmacology , ran GTP-Binding Protein/metabolism , ran GTP-Binding Protein/pharmacology , Hippocampus/metabolism , Ischemia/metabolism , Oxidative Stress , Brain Ischemia/metabolism , Apoptosis , Gene Products, tat/genetics , Gene Products, tat/metabolism , Gene Products, tat/pharmacology , Disease Models, Animal , Brain Injuries/metabolism , Neuroprotective Agents/pharmacology
4.
Parasitol Res ; 121(11): 3229-3241, 2022 Nov.
Article En | MEDLINE | ID: mdl-36056961

Tyrosine aminotransferase is a well-characterized enzyme in the Leishmania parasite, but the role of TAT in the parasite functioning remains largely unknown. In this study, we attempt to gain a better understanding of the enzyme's role in the parasite by gene knockout and overexpression of the TAT gene. The overexpression of TAT protein was well tolerated by the parasites in two independent repeats. Single knockout of TAT gene by homologous recombination, LdTAT+/- displayed distinct retardation in the proliferation rates and entered the death phase immediately. Morphology of LdTAT+/- parasites had important structural defects as they rounded up with elongated flagella. Gene regulation studies suggested the upregulation of key apoptotic and redox metabolism genes in LdTAT+/-. Moreover, LdTAT+/- cells accumulated higher ROS, thiols, intracellular Ca2+ concentrations, and mitochondrial membrane depolarization signifying the onset of apoptosis. Tocopherol levels were reduced by 50% in LdTAT+/- suggesting the involvement of TAT in tocopherol biosynthesis in the parasite. Overall, our results provide the first evidence that gene knockout of TAT results in apoptosis and that TAT is required for the survival and viability of Leishmania donovani.


Leishmania donovani , Parasites , Animals , Gene Products, tat/genetics , Gene Products, tat/metabolism , Homeostasis , Homologous Recombination , Oxidation-Reduction , Parasites/metabolism , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism , Tocopherols/metabolism , Tyrosine Transaminase/chemistry , Tyrosine Transaminase/genetics , Tyrosine Transaminase/metabolism
5.
Int J Nanomedicine ; 17: 3269-3286, 2022.
Article En | MEDLINE | ID: mdl-35924260

Background: Naringin is a naturally occurring flavanone that promotes osteogenesis. Owing to the high lipophilicity, poor in vivo bioavailability, and extensive metabolic alteration upon administration, the clinical efficacy of naringin is understudied. Additionally, information on the molecular mechanism by which it promotes osteogenesis is limited. Methods: In this study, we prepared TAT & RGD peptide-modified naringin-loaded nanoparticles (TAT-RGD-NAR-NPs), evaluated their potency on the osteogenic differentiation of human dental pulp stem cells (hDPSCs), and studied its mechanism of action through metabolomic analysis. Results: The particle size and zeta potential of TAT-RGD-NAR-NPs were 160.70±2.05 mm and -20.77±0.47mV, respectively. The result of cell uptake assay showed that TAT-RGD-NAR-NPs could effectively enter hDPSCs. TAT-RGD-NAR-NPs had a more significant effect on cell proliferation and osteogenic differentiation promotion. Furthermore, in metabolomic analysis, naringin particles showed a strong influence on the glycerophospholipid metabolism pathway of hDPSCs. Specifically, it upregulated the expression of PLA2G3 and PLA2G1B (two isozymes of phospholipase A2, PLA2), increased the biosynthesis of lysophosphatidic acid (LPA). Conclusion: These results suggested that TAT-RGD-NPs might be used for transporting naringin to hDPSCs for modulating stem cell osteogenic differentiation. The metabolomic analysis was used for the first time to elucidate the mechanism by which naringin promotes hDPSCs osteogenesis by upregulating PLA2G3 and PLA2G1B.


Flavanones , Nanoparticles , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Dental Pulp , Flavanones/pharmacology , Gene Products, tat/genetics , Group IB Phospholipases A2/metabolism , Group III Phospholipases A2/metabolism , Humans , Liposomes , Oligopeptides/metabolism , Osteogenesis , Stem Cells
6.
Int J Biol Macromol ; 209(Pt A): 972-983, 2022 Jun 01.
Article En | MEDLINE | ID: mdl-35460749

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.


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
7.
Biochem J ; 479(3): 259-272, 2022 02 11.
Article En | MEDLINE | ID: mdl-35015082

Murine cytomegalovirus protein M45 contains a RIP homotypic interaction motif (RHIM) that is sufficient to confer protection of infected cells against necroptotic cell death. Mechanistically, the N-terminal region of M45 drives rapid self-assembly into homo-oligomeric amyloid fibrils, and interacts with the endogenous RHIM domains of receptor-interacting serine/threonine protein kinases (RIPK) 1, RIPK3, Z-DNA-binding protein 1, and Toll/interleukin-1 receptor domain-containing adaptor-inducing interferon-ß. Remarkably, all four aforementioned mammalian proteins harbouring such a RHIM domain are key components of inflammatory signalling and regulated cell death (RCD) processes. Immunogenic cell death by regulated necrosis causes extensive tissue damage in a wide range of diseases, including ischaemia reperfusion injury, myocardial infarction, sepsis, stroke, and solid organ transplantation. To harness the cell death suppression properties of M45 protein in a therapeutically usable manner, we developed a synthetic peptide encompassing only the RHIM domain of M45. To trigger delivery of RHIM into target cells, we fused the transactivator protein transduction domain of human immunodeficiency virus 1 to the N-terminus of the peptide. The fused peptide could efficiently penetrate eukaryotic cells, but unexpectedly it eradicated or destroyed all tested cancer cell lines and primary cells irrespective of species without further stimulus through a necrosis-like cell death. Typical inhibitors of different forms of RCD cannot impede this process, which appears to involve a direct disruption of biomembranes. Nevertheless, our finding has potential clinical relevance; reliable induction of a necrotic form of cell death distinct from all known forms of RCD may offer a novel therapeutic approach to combat resistant tumour cells.


Gene Products, tat/chemistry , Gene Products, tat/metabolism , Protein Domains , Recombinant Fusion Proteins/metabolism , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism , Signal Transduction/genetics , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Amyloid/metabolism , Animals , Gene Products, tat/genetics , HIV-1/chemistry , HT29 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Necroptosis/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ribonucleotide Reductases/genetics , U937 Cells , Viral Proteins/genetics
8.
Biochem Biophys Res Commun ; 586: 63-67, 2022 01 01.
Article En | MEDLINE | ID: mdl-34826702

Although cell-penetrating peptides such as the HIV-derived TAT peptide have been used as tools for the intracellular delivery of therapeutic peptides and proteins, a problem persists: the endosomal escape efficiency is low. Previously, we found that the fusogenic peptide S19, derived from the human protein syncytin-1, enhance the endosomal escape efficiency of proteins that incorporated by endocytosis via TAT. In this study, we first performed Ala-scanning mutagenesis of S19, and found that all Ile, Val, Leu and Phe with high ß-sheet forming propensities in S19 are important for the intracellular uptake of S19-TAT-fused proteins. In a secondary structure analysis of the mutated S19-TAT peptides in the presence of liposomes mimicking late endosomes (LEs), the CD spectra of V3A and I4A mutants with low uptake activity showed the appearance of an α-helix structure, whereas the mutant G5A retained both the uptake activity and the ß-structure. In addition, we investigated the appropriate linking position and order of the S19 and TAT peptides to a cargo protein including an apoptosis-induced peptide and found that both the previous C-terminal S19-TAT tag and the N-terminal TAT-S19 tag promote the cytoplasmic delivery of the fusion protein. These results and previous results suggest that the interaction of TAT with the LE membrane causes a structural change in S19 from a random coil to a ß-strand and that the subsequent parallel ß-sheet formation between two S19 peptides may promote adjacent TAT dimerization, resulting in endosomal escape from the LE membrane.


Cell Membrane/metabolism , Gene Products, env/metabolism , Gene Products, tat/metabolism , Peptides/metabolism , Plasmids/metabolism , Pregnancy Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Amino Acid Substitution , Cell Line, Tumor , Cell Membrane Permeability , Endosomes/chemistry , Endosomes/metabolism , Gene Expression , Gene Products, env/genetics , Gene Products, tat/genetics , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Liposomes/chemistry , Liposomes/metabolism , Optical Imaging , Peptides/genetics , Plasmids/chemistry , Pregnancy Proteins/genetics , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Transport , Recombinant Fusion Proteins/genetics , Transduction, Genetic
9.
PLoS One ; 16(12): e0260010, 2021.
Article En | MEDLINE | ID: mdl-34855793

Little is known about how specific individual viral lineages replicating systemically during acute Human Immunodeficiency Virus or Simian Immunodeficiency Virus (HIV/SIV) infection persist into chronic infection. In this study, we use molecularly barcoded SIV (SIVmac239M) to track distinct viral lineages for 12 weeks after intravenous (IV) or intrarectal (IR) challenge in macaques. Two Mafa-A1*063+ cynomolgus macaques (Macaca fascicularis, CM) were challenged IV, and two Mamu-A1*001+ rhesus macaques (Macaca mulatta, RM) were challenged IR with 200,000 Infectious Units (IU) of SIVmac239M. We sequenced the molecular barcode of SIVmac239M from all animals over the 12 weeks of the study to characterize the diversity and persistence of virus lineages. During the first three weeks post-infection, we found ~70-560 times more unique viral lineages circulating in the animals challenged IV compared to those challenged IR, which is consistent with the hypothesis that the challenge route is the primary driver restricting the transmission of individual viral lineages. We also characterized the sequences of T cell epitopes targeted during acute SIV infection, and found that the emergence of escape variants in acutely targeted epitopes can occur on multiple virus templates simultaneously, but that elimination of some of these templates is likely a consequence of additional host factors. These data imply that virus lineages present during acute infection can still be eliminated from the systemic virus population even after initial selection.


Mucous Membrane/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/growth & development , Animals , Epitopes/immunology , Female , Gene Products, tat/genetics , Injections, Intravenous , Macaca fascicularis/immunology , Macaca fascicularis/virology , Macaca mulatta/immunology , Macaca mulatta/virology , Mucous Membrane/immunology , Mutation , RNA, Viral/blood , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , T-Lymphocytes/immunology , T-Lymphocytes/virology , Viral Load , Viremia/immunology , Viremia/virology
10.
PLoS One ; 16(9): e0256715, 2021.
Article En | MEDLINE | ID: mdl-34499687

The twin-arginine translocation (Tat) pathway transports folded proteins across energetic membranes. Numerous Tat substrates contain co-factors that are inserted before transport with the assistance of redox enzyme maturation proteins (REMPs), which bind to the signal peptide of precursor proteins. How signal peptides are transferred from a REMP to a binding site on the Tat receptor complex remains unknown. Since the signal peptide mediates both interactions, possibilities include: i) a coordinated hand-off mechanism; or ii) a diffusional search after REMP dissociation. We investigated the binding interaction between substrates containing the TorA signal peptide (spTorA) and its cognate REMP, TorD, and the effect of TorD on the in vitro transport of such substrates. We found that Escherichia coli TorD is predominantly a monomer at low micromolar concentrations (dimerization KD > 50 µM), and this monomer binds reversibly to spTorA (KD ≈ 1 µM). While TorD binds to membranes (KD ≈ 100 nM), it has no apparent affinity for Tat translocons and it inhibits binding of a precursor substrate to the membrane. TorD has a minimal effect on substrate transport by the Tat system, being mildly inhibitory at high concentrations. These data are consistent with a model in which the REMP-bound signal peptide is shielded from recognition by the Tat translocon, and spontaneous dissociation of the REMP allows the substrate to engage the Tat machinery. Thus, the REMP does not assist with targeting to the Tat translocon, but rather temporarily shields the signal peptide.


Escherichia coli Proteins/genetics , Gene Products, tat/genetics , Molecular Chaperones/genetics , Oxidoreductases, N-Demethylating/genetics , Twin-Arginine-Translocation System/genetics , Binding Sites/genetics , Escherichia coli/genetics , Protein Binding/genetics , Protein Sorting Signals/genetics , Protein Transport/genetics , Substrate Specificity
11.
Molecules ; 26(11)2021 Jun 03.
Article En | MEDLINE | ID: mdl-34205205

Total body irradiation is a standard procedure of bone marrow transplantation (BMT) which causes a rapid increase in reactive oxygen species (ROS) in the bone marrow microenvironment during BMT. The increase in ROS reduces the engraftment ability of donor cells, thereby affecting the bone marrow recovery of recipients after BMT. In the early weeks following transplantation, recipients are at high risk of severe infection due to weakened hematopoiesis. Thus, it is imperative to improve engraftment capacity and accelerate bone marrow recovery in BMT recipients. In this study, we constructed recombinant copper/zinc superoxide dismutase 1 (SOD1) fused with the cell-penetrating peptide (CPP), the trans-activator of transcription (Tat), and showed that this fusion protein has penetrating ability and antioxidant activity in both RAW264.7 cells and bone marrow cells in vitro. Furthermore, irradiated mice transplanted with SOD1-Tat-treated total bone marrow donor cells showed an increase in total bone marrow engraftment capacity two weeks after transplantation. This study explored an innovative method for enhancing engraftment efficiency and highlights the potential of CPP-SOD1 in ROS manipulation during BMT.


Antioxidants/pharmacology , Bone Marrow Cells/cytology , Cell-Penetrating Peptides/genetics , Gene Products, tat/genetics , Recombinant Fusion Proteins/pharmacology , Superoxide Dismutase-1/genetics , Animals , Antioxidants/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Transplantation , Cell-Penetrating Peptides/metabolism , Cells, Cultured , Gene Products, tat/metabolism , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species , Recombinant Fusion Proteins/metabolism , Superoxide Dismutase-1/metabolism , Whole-Body Irradiation
12.
BMB Rep ; 54(9): 458-463, 2021 Sep.
Article En | MEDLINE | ID: mdl-34120676

Cytokines activate inflammatory signals and are major mediators in progressive ß-cell damage, which leads to type 1 diabetes mellitus. We recently showed that the cell-permeable Tat-CIAPIN1 fusion protein inhibits neuronal cell death induced by oxidative stress. However, how the Tat-CIAPIN1 protein affects cytokine-induced ß-cell damage has not been investigated yet. Thus, we assessed whether the Tat-CIAPIN1 protein can protect RINm5F ß-cells against cytokine-induced cytotoxicity. In cytokine-exposed RINm5F ß-cells, the transduced Tat-CIAPIN1 protein elevated cell survivals and reduced reactive oxygen species (ROS) and DNA fragmentation levels. The Tat-CIAPIN1 protein reduced mitogen-activated protein kinases (MAPKs) and NF-κB activation levels and elevated Bcl-2 protein, whereas Bax and cleaved Caspase-3 proteins were decreased by this fusion protein. Thus, the protection of RINm5F ß-cells by the Tat-CIAPIN1 protein against cytokine-induced cytotoxicity can suggest that the Tat-CIAPIN1 protein might be used as a therapeutic inhibitor against RINm5F ß-cell damage. [BMB Reports 2021; 54(9): 458-463].


Apoptosis/drug effects , Cytokines/pharmacology , Gene Products, tat/genetics , Intracellular Signaling Peptides and Proteins/genetics , Recombinant Fusion Proteins/metabolism , Caspase 3/metabolism , Cell Line , DNA Fragmentation/drug effects , Gene Products, tat/metabolism , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/genetics , Signal Transduction/drug effects
13.
mBio ; 12(3): e0130221, 2021 06 29.
Article En | MEDLINE | ID: mdl-34154411

The cell envelope of Gram-negative bacteria consists of two membranes surrounding the periplasm and peptidoglycan layer. ß-Lactam antibiotics target the periplasmic penicillin-binding proteins that synthesize peptidoglycan, resulting in cell death. The primary means by which bacterial species resist the effects of ß-lactam drugs is to populate the periplasmic space with ß-lactamases. Resistance to ß-lactam drugs is spread by lateral transfer of genes encoding ß-lactamases from one species of bacteria to another. However, the resistance phenotype depends in turn on these "alien" protein sequences being recognized and exported across the cytoplasmic membrane by either the Sec or Tat protein translocation machinery of the new bacterial host. Here, we examine BKC-1, a carbapenemase from an unknown bacterial source that has been identified in a single clinical isolate of Klebsiella pneumoniae. BKC-1 was shown to be located in the periplasm, and functional in both K. pneumoniae and Escherichia coli. Sequence analysis revealed the presence of an unusual signal peptide with a twin arginine motif and a duplicated hydrophobic region. Biochemical assays showed this signal peptide directs BKC-1 for translocation by both Sec and Tat translocons. This is one of the few descriptions of a periplasmic protein that is functionally translocated by both export pathways in the same organism, and we suggest it represents a snapshot of evolution for a ß-lactamase adapting to functionality in a new host. IMPORTANCE Bacteria can readily acquire plasmids via lateral gene transfer (LGT). These plasmids can carry genes for virulence and antimicrobial resistance (AMR). Of growing concern are LGT events that spread ß-lactamases, particularly carbapenemases, and it is important to understand what limits this spread. This study provides insight into the sequence features of BKC-1 that exemplify the limitations on the successful biogenesis of ß-lactamases, which is one factor limiting the spread of AMR phenotypes by LGT. With a very simple evolutionary adaptation, BKC-1 could become a more effective carbapenemase, underscoring the need to understand the evolution, adaptability, and functional assessment of newly reported ß-lactamases rapidly and thoroughly.


Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Products, tat/genetics , Klebsiella pneumoniae/genetics , SEC Translocation Channels/genetics , beta-Lactamases/genetics , beta-Lactamases/metabolism , Anti-Bacterial Agents/pharmacology , Biological Transport , Escherichia coli/genetics , Humans , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/enzymology , Microbial Sensitivity Tests , Periplasm/metabolism , beta-Lactams/pharmacology
14.
Cells ; 9(8)2020 08 03.
Article En | MEDLINE | ID: mdl-32756411

Cannabinoid receptor-interacting protein 1a (CRIP1a) binds to the C-terminal domain of cannabinoid 1 receptor (CB1R) and regulates CB1R activities. In this study, we made Tat-CRIP1a fusion proteins to enhance CRIP1a penetration into neurons and brain and to evaluate the function of CRIP1a in neuroprotection following oxidative stress in HT22 hippocampal cells and transient forebrain ischemia in gerbils. Purified exogenous Tat-CRIP1a was penetrated into HT22 cells in a time and concentration-dependent manner and prevented H2O2-induced reactive oxygen species formation, DNA fragmentation, and cell damage. Tat-CRIP1a fusion protein also ameliorated the reduction of 14-3-3η expression by H2O2 treatment in HT22 cells. Ischemia-reperfusion damage caused motor hyperactivity in the open field test of gerbils; however, the treatment of Tat-CRIP1a significantly reduced hyperactivity 1 day after ischemia. Four days after ischemia, the administration of Tat-CRIP1a restored the loss of pyramidal neurons and decreased reactive astrocytosis and microgliosis induced by ischemic damage in the hippocampal cornu Ammonis (CA)1 region. Ischemic damage decreased 14-3-3η expression in all hippocampal sub-regions 4 days after ischemia; however, the treatment of Tat-CRIP1 ameliorated the reduction of 14-3-3η expression. These results suggest that Tat-CRIP1a attenuates neuronal damage and hyperactivity induced by ischemic damage, and it restores normal expression levels of 14-3-3η protein in the hippocampus.


14-3-3 Proteins/genetics , Gene Products, tat/genetics , Ischemia/pathology , Membrane Proteins/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , 14-3-3 Proteins/metabolism , Animals , Cell Death/drug effects , Cell Line , DNA Damage , Gerbillinae , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Ischemia/drug therapy , Ischemia/metabolism , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Motor Activity/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology
15.
Int J Mol Sci ; 21(8)2020 Apr 11.
Article En | MEDLINE | ID: mdl-32290442

Reactive oxygen species (ROS) is major risk factor in neuronal diseases including ischemia. Although biliverdin reductase A (BLVRA) plays a pivotal role in cell survival via its antioxidant function, its role in hippocampal neuronal (HT-22) cells and animal ischemic injury is not clearly understood yet. In this study, the effects of transducible fusion protein Tat-BLVRA on H2O2-induced HT-22 cell death and in an animal ischemia model were investigated. Transduced Tat-BLVRA markedly inhibited cell death, DNA fragmentation, and generation of ROS. Transduced Tat-BLVRA inhibited the apoptosis and mitogen activated protein kinase (MAPK) signaling pathway and it passed through the blood-brain barrier (BBB) and significantly prevented hippocampal cell death in an ischemic model. These results suggest that Tat-BLVRA provides a possibility as a therapeutic molecule for ischemia.


Apoptosis/drug effects , Gene Products, tat , MAP Kinase Signaling System/drug effects , Oxidative Stress/drug effects , Oxidoreductases Acting on CH-CH Group Donors , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Brain Ischemia/drug therapy , Brain Ischemia/etiology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Line , Disease Models, Animal , Gene Products, tat/genetics , Gerbillinae , Hydrogen Peroxide/metabolism , Male , Neuroprotective Agents/pharmacology , Oxidoreductases Acting on CH-CH Group Donors/genetics , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/genetics
16.
FASEB J ; 34(3): 4147-4162, 2020 03.
Article En | MEDLINE | ID: mdl-31950548

HIV-1 Tat is essential for HIV-1 replication and appears to play an important role in the pathogenesis of HIV-associated neurological complications. Secreted from infected or transfected cells, Tat has the extraordinary ability to cross the plasma membrane. In the brain, Tat can be taken up by CNS cells via receptor-mediated endocytosis. Following endocytosis and its internalization into endolysosomes, Tat must be released in order for it to activate the HIV-1 LTR promoter and facilitate HIV-1 viral replication in the nucleus. However, the underlying mechanisms whereby Tat escapes endolysosomes remain unclear. Because Tat disrupts intracellular calcium homeostasis, we investigated the involvement of calcium in Tat endolysosome escape and subsequent LTR transactivation. We demonstrated that chelating endolysosome calcium with high-affinity rhodamine-dextran or chelating cytosolic calcium with BAPTA-AM attenuated Tat endolysosome escape and LTR transactivation. Significantly, we demonstrated that pharmacologically blocking and knocking down the endolysosome-resident two-pore channels (TPCs) attenuated Tat endolysosome escape and LTR transactivation. This calcium-mediated effect appears to be selective for TPCs because knocking down TRPML1 calcium channels was without effect. Our findings suggest that calcium released from TPCs is involved in Tat endolysosome escape and subsequent LTR transactivation. TPCs might represent a novel therapeutic target against HIV-1 infection and HIV-associated neurological complications.


Calcium/metabolism , Gene Products, tat/metabolism , Cell Line, Tumor , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/physiology , Gene Products, tat/genetics , HIV Long Terminal Repeat/genetics , HIV Long Terminal Repeat/physiology , HIV-1/metabolism , Humans , Immunoblotting , Lysosomes/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Virus Replication/genetics , Virus Replication/physiology
17.
Int J Nanomedicine ; 14: 8627-8645, 2019.
Article En | MEDLINE | ID: mdl-31806961

BACKGROUND AND PURPOSE: Systemic lupus erythematous (SLE) is an autoimmune disease caused by many factors. Lupus nephritis (LN) is a common complication of SLE and represents a major cause of morbidity and mortality. Previous studies have shown the advantages of multi-targeted therapy for LN and that TLR4 signaling is a target of anti-LN drugs. High-mobility group box 1 (HMGB1), a nuclear protein with a proinflammatory cytokine activity, binds specifically to TLR4 to induce inflammation. We aimed to develop PEGylated TAT peptide-cationic liposomes (TAT-CLs) to deliver anti-HMGB1 siRNA and dihydroartemisinin (DHA) to increase LN therapeutic efficiency and explore their treatment mechanism. METHODS: We constructed the TAT-CLs-DHA/siRNA delivery system using the thin film hydration method. The uptake and localization of Cy3-labeled siRNA were detected by confocal microscopy and flow cytometry. MTT assays were used to detect glomerular mesangial cell proliferation. Real-time PCR, Western blot analysis, and ELISA evaluated the anti-inflammatory mechanism of TAT-CLs-DHA/siRNA. RESULTS: We constructed the TAT-CLs-DHA/siRNA delivery system measuring approximately 140 nm with superior storage and serum stabilities. In vitro, it showed significantly greater uptake compared with unmodified liposomes and significant inhibition of glomerular mesangial cell proliferation. TAT-CLs-DHA/siRNA inhibited NF-κB activation in a concentration-dependent manner. Real-time PCR and Western blot analysis showed that TAT-CLs-DHA/siRNA downregulated expression of HMGB1 mRNA and protein. TAT-CLs-DHA/siRNA markedly diminished Toll-like receptor 4 (TLR4) expression and subsequent activation of MyD88, IRAK4, and NF-κB. CONCLUSION: TAT-CLs-DHA/siRNA may have the potential for treatment of inflammatory diseases such as LN mediated by the TLR4 signaling pathway.


Artemisinins/administration & dosage , Gene Products, tat/genetics , HMGB1 Protein/genetics , Liposomes/administration & dosage , Lupus Nephritis/therapy , RNA, Small Interfering/administration & dosage , Toll-Like Receptor 4/metabolism , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Gene Expression Regulation/drug effects , Humans , Liposomes/chemistry , Liposomes/pharmacology , Lupus Nephritis/metabolism , NF-kappa B/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Int J Mol Sci ; 20(19)2019 Oct 05.
Article En | MEDLINE | ID: mdl-31590403

G-protein-coupled receptors associate into dimers/oligomers whose function is not well understood. One approach to investigate this issue is to challenge oligomerization by peptides replicating transmembrane domains and to study their effect on receptor functionality. The disruptor peptides are typically delivered by means of cell-penetrating vectors such as the human immunodeficiency virus (HIV) transcription trans-activation protein Tat. In this paper we report a cyclic, Tat-like peptide that significantly improves its linear analogue in targeting interreceptor sequences in the transmembrane space. The same cyclic Tat-like vector fused to a transmembrane region not involved in receptor oligomerization was totally ineffective. Besides higher efficacy, the cyclic version has enhanced proteolytic stability, as shown by trypsin digestion experiments.


Gene Products, tat/metabolism , Peptides, Cyclic/metabolism , Receptor, Adenosine A2A/metabolism , Gene Products, tat/genetics , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Peptides, Cyclic/genetics , Protein Binding , Protein Multimerization , Protein Stability
19.
EcoSal Plus ; 8(2)2019 06.
Article En | MEDLINE | ID: mdl-31215506

The Tat pathway for protein translocation across bacterial membranes stands out for its selective handling of fully folded cargo proteins. In this review, we provide a comprehensive summary of our current understanding of the different known Tat components, their assembly into different complexes, and their specific roles in the protein translocation process. In particular, this overview focuses on the Gram-negative bacterium Escherichia coli and the Gram-positive bacterium Bacillus subtilis. Using these organisms as examples, we discuss structural features of Tat complexes alongside mechanistic models that allow for the Tat pathway's unique protein proofreading and transport capabilities. Finally, we highlight recent advances in exploiting the Tat pathway for biotechnological benefit, the production of high-value pharmaceutical proteins.


Arginine/metabolism , Bacillus subtilis/metabolism , Escherichia coli/metabolism , Gene Products, tat/metabolism , Protein Transport , Bacillus subtilis/genetics , Escherichia coli/genetics , Gene Products, tat/genetics , Metabolic Networks and Pathways , Protein Sorting Signals
20.
Free Radic Biol Med ; 135: 68-78, 2019 05 01.
Article En | MEDLINE | ID: mdl-30818058

Cytokine-induced apoptosis inhibitor 1 (CIAPIN1) protein is widely expressed in the brain and it is known that this protein is involved in cell survival including dopaminergic neuronal cells. Oxidative stress is known as one of the major causes of degenerative diseases including ischemia. In this study, we investigated the effect of CIAPIN1 protein on hippocampal neuronal (HT-22) cell damage induced by hydrogen peroxide (H2O2) and in an animal model of ischemia using Tat-CIAPIN1 fusion protein which can transduce into cells. Tat-CIAPIN1 protein transduced into HT-22 cells and significantly inhibited cell death, DNA fragmentation, and reactive oxygen species (ROS) generation. Also, Tat-CIAPIN1 protein enhances cell survival via the regulation of Akt, MAPK, NF-κB and apoptotic signaling pathways in the H2O2 treated cells. In an ischemic animal model, Tat-CIAPIN1 protein transduced into the brain and protected neuronal cell death of hippocampal CA1 region induced by ischemic insult. In conclusion, we demonstrated that Tat-CIAPIN1 protein has protective effects against hippocampal neuronal cell damage induced by ischemic injury, suggesting that Tat-CIAPIN1 protein may provide a potential therapeutic agent for ischemia.


Brain Ischemia/genetics , Gene Products, tat/genetics , Intracellular Signaling Peptides and Proteins/genetics , Neurodegenerative Diseases/genetics , Animals , Apoptosis/genetics , Brain/drug effects , Brain/metabolism , Brain Ischemia/pathology , Brain Ischemia/prevention & control , Cell Survival/drug effects , DNA Fragmentation/drug effects , Disease Models, Animal , Genetic Therapy , Hippocampus/metabolism , Hippocampus/pathology , Humans , Hydrogen Peroxide/metabolism , Intracellular Signaling Peptides and Proteins/therapeutic use , Mice , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/prevention & control , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
...