Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 313(4): G342-G352, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28705805

ABSTRACT

There is a lack of tools that selectively target vagal afferent neurons (VAN) innervating the gut. We use saporin (SAP), a potent neurotoxin, conjugated to the gastronintestinal (GI) hormone cholecystokinin (CCK-SAP) injected into the nodose ganglia (NG) of male Wistar rats to specifically ablate GI-VAN. We report that CCK-SAP ablates a subpopulation of VAN in culture. In vivo, CCK-SAP injection into the NG reduces VAN innervating the mucosal and muscular layers of the stomach and small intestine but not the colon, while leaving vagal efferent neurons intact. CCK-SAP abolishes feeding-induced c-Fos in the NTS, as well as satiation by CCK or glucagon like peptide-1 (GLP-1). CCK-SAP in the NG of mice also abolishes CCK-induced satiation. Therefore, we provide multiple lines of evidence that injection of CCK-SAP in NG is a novel selective vagal deafferentation technique of the upper GI tract that works in multiple vertebrate models. This method provides improved tissue specificity and superior separation of afferent and efferent signaling compared with vagotomy, capsaicin, and subdiaphragmatic deafferentation.NEW & NOTEWORTHY We develop a new method that allows targeted lesioning of vagal afferent neurons that innervate the upper GI tract while sparing vagal efferent neurons. This reliable approach provides superior tissue specificity and selectivity for vagal afferent over efferent targeting than traditional approaches. It can be used to address questions about the role of gut to brain signaling in physiological and pathophysiological conditions.


Subject(s)
Afferent Pathways/drug effects , Autonomic Denervation/methods , Gastrointestinal Tract/drug effects , Nerve Block/methods , Ribosome Inactivating Proteins, Type 1/administration & dosage , Vagus Nerve/drug effects , Afferent Pathways/physiology , Animals , Gastrointestinal Tract/physiology , Male , Neurotoxins/administration & dosage , Neurotoxins/pharmacology , Rats , Rats, Wistar , Saporins , Treatment Outcome , Vagus Nerve/physiology
2.
BMC Neurosci ; 18(1): 29, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28264667

ABSTRACT

BACKGROUND: The possibility of using deep brain stimulation (DBS) for memory enhancement has recently been reported, but the precise underlying mechanisms of its effects remain unknown. Our previous study suggested that spatial memory improvement by medial septum (MS)-DBS may be associated with cholinergic regulation and neurogenesis. However, the affected stage of memory could not be distinguished because the stimulation was delivered during the execution of all memory processes. Therefore, this study was performed to determine the stage of memory affected by MS-DBS. Rats were administered 192 IgG-saporin to lesion cholinergic neurons. Stimulation was delivered at different times in different groups of rats: 5 days before the Morris water maze test (pre-stimulation), 5 days during the training phase of the Morris water maze test (training-stimulation), and 2 h before the Morris water maze probe test (probe-stimulation). A fourth group of rats was lesioned but received no stimulation. These four groups were compared with a normal (control) group. RESULTS: The most effective memory restoration occurred in the pre-stimulation group. Moreover, the pre-stimulation group exhibited better recall of the platform position than the other stimulation groups. An increase in the level of brain derived neurotrophic factor (BDNF) was observed in the pre-stimulation group; this increase was maintained for 1 week. However, acetylcholinesterase activity in the pre-stimulation group was not significantly different from the lesion group. CONCLUSION: Memory impairment due to cholinergic denervation can be improved by DBS. The improvement is significantly correlated with the up-regulation of BDNF expression and neurogenesis. Based on the results of this study, the use of MS-DBS during the early stage of disease may restore spatial memory impairment.


Subject(s)
Deep Brain Stimulation/methods , Maze Learning/physiology , Septal Nuclei/physiology , Spatial Memory/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Brain-Derived Neurotrophic Factor/metabolism , Choline O-Acetyltransferase/metabolism , Cholinergic Agents/administration & dosage , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Glutamate Decarboxylase/metabolism , Male , Neurogenesis , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1/administration & dosage , Saporins , Septal Nuclei/drug effects , Septal Nuclei/metabolism
3.
Acta Pharmacol Sin ; 38(6): 897-906, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28414205

ABSTRACT

Therapeutically potent macromolecular drugs have shown great promise for overcoming the limitations of small-molecule anti-cancer drugs. But tumor cell-selective intracellular delivery of the macromolecules remains a major hurdle for their successful clinical application. To overcome this challenge, we engineered a novel genetic fusion protein (F3-Gel) that composed of F3 peptide, a tumor-homing peptide, and gelonin, a plant-derived ribosome-inactivating protein (RIP), and then evaluated its anti-cancer activity in vitro and in vivo. The F3-Gel-encoding gene was synthesized by genetic recombination, and F3-Gel was successfully expressed in E coli. The anti-cancer activity of the produced F3-Gel was evaluated by various in vitro assays, which revealed that F3-Gel maintained equipotent protein synthesis inhibition activity (IC50=11 pmol/L) as unmodified gelonin (IC50=10 pmol/L). Furthermore, F3-Gel displayed enhanced cellular uptake into cancer cells (U87 MG, HeLa, LnCaP and 9L) than noncancerous cells (293 HEK and SVGp12). Compared with gelonin, F3-Gel exerted significantly higher cytotoxicity against these cancer cells. F3-Gel displayed significantly greater inhibition of protein translation in U87 MG cells: F3-Gel (0.5 µmol/L) was able to reduce the protein level to less than 50%, while gelonin (1 µmol/L) did not affect the intracellular protein level. In a U87 MG xenograft tumor-bearing mouse model, F3-Gel was accumulated in the tumor site at much higher levels and maintained for a prolonged time compared with gelonin. Administration of F3-Gel (0.5, 0.75 mol/kg, iv) caused 36% and 66%, respectively, inhibition of tumor growth in U87 MG xenograft mice, suggesting that it is a promising candidate drug for cancer treatment. Furthermore, this study demonstrates that fusion of F3 peptide to a potent macromolecule could provides an effective method for targeting tumors and eventually could improve their druggability.


Subject(s)
Antineoplastic Agents/pharmacology , Peptides/pharmacology , Ribosome Inactivating Proteins, Type 1/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Gels/administration & dosage , Gels/chemistry , Gels/pharmacology , Humans , Injections, Intravenous , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Male , Mice , Mice, Nude , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/drug therapy , Peptides/administration & dosage , Peptides/chemistry , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/chemistry , Structure-Activity Relationship
4.
Biochim Biophys Acta ; 1840(6): 1657-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24361614

ABSTRACT

BACKGROUND: Gliomas have been termed recurrent cancers due to their highly aggressive nature. Their tendency to infiltrate and metastasize has posed significant roadblocks to in attaining fool proof treatment solutions. An initiative to curb such a scenario was successfully demonstrated in vitro, utilizing a multi-conceptual gold nanoparticle based photo-thermal and drug combination therapy. METHODS: Gold nanoparticles (Au NPs) were synthesized with a highly environmentally benign process. The Au NPs were PEGylated and conjugated with folate and transferrin antibody to achieve a dual targeted nano-formulation directed towards gliomas. Curcin, a type 1 ribosome inactivating protein, was attached to the Au NPs as the drug candidate, and its multifarious toxic aspects analyzed in vitro. NIR photo-thermal properties of the Au nano-conjugates were studied to selectively ablate the glioma cancer colonies. RESULTS: Highly cyto-compatible, 10-15nm Au NP conjugates were synthesized with pronounced specificity towards gliomas. Curcin was successfully conjugated to the Au NPs with pH responsive drug release. Prominent toxic aspects of curcin, such as ROS generation, mitochondrial and cytoskeletal destabilization were witnessed. Excellent photo-thermal ablation properties of gold nanoparticles were utilized to completely disrupt the cancer colonies with significant precision. CONCLUSION: The multifunctional nanoconjugate projects its competence in imparting complete arrest of the future proliferation or migration of the cancer mass. GENERAL SIGNIFICANCE: With multifunctionality the essence of nanomedicine in recent years, the present nanoconjugate highlights itself as a viable option for a multimodal treatment option for brain cancers and the like.


Subject(s)
Brain Neoplasms/drug therapy , Gold/administration & dosage , Metal Nanoparticles/administration & dosage , Ribosome Inactivating Proteins, Type 1/administration & dosage , Cells, Cultured , Humans , Reactive Oxygen Species/metabolism , Trichothecenes
5.
Mol Pain ; 11: 54, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26353788

ABSTRACT

BACKGROUND: Non-peptidergic nociceptive neurons are a sub-population of small diameter primary sensory neurons that comprise approximately 50 % of the C fiber population. Together with the peptidergic sub-population, they transmit nociceptive information from the periphery to the superficial dorsal horn of the spinal cord. Despite the numerous studies investigating the role of the non-peptidergic primary afferents, their role in normal nociception and in pain remains poorly understood. Our lab has previously demonstrated that, in rat models of neuropathic and inflammatory pain, there is a de novo expression of substance P receptors (NK-1r) by lamina I pyramidal projection neurons, a neuronal population that normally does not express these receptors. RESULTS: In this study, we used a ribosomal toxin, saporin, conjugated to the lectin IB4 to selectively ablate the non-peptidergic nociceptive C fibers, to investigate if the loss of these fibers was enough to induce a change in NK-1r expression by lamina I projection neurons. IB4-saporin treatment led to the permanent ablation of the IB4-positive afferents but also to a small non-significant reduction in CGRP-positive afferents. An overall increase in immunoreactivity for the NK-1r was observed in lamina I projection neurons, however, the lack of non-peptidergic afferents did not increase the number of lamina I pyramidal projection neurons immunoreactive for the receptor. CONCLUSIONS: Our results demonstrate that the deletion of the non-peptidergic afferents, at the L4-L5 spinal levels, is not sufficient to trigger the de novo expression of NK-1r by projection pyramidal neurons but increases the expression of NK-1r in fusiform and multipolar projection neurons. Furthermore, our data suggest that a neuropathic component is essential to trigger the expression of NK-1r by pyramidal neurons.


Subject(s)
Nerve Fibers, Unmyelinated/metabolism , Peptides/metabolism , Posterior Horn Cells/metabolism , Animals , Behavior, Animal/drug effects , Injections , Lectins/administration & dosage , Lectins/pharmacology , Male , Microscopy, Confocal , Nerve Fibers, Unmyelinated/drug effects , Posterior Horn Cells/drug effects , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins
6.
Am J Physiol Regul Integr Comp Physiol ; 309(4): R358-67, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26062632

ABSTRACT

Both lateral hypothalamic orexinergic neurons and hindbrain catecholaminergic neurons contribute to control of feeding behavior. Orexin fibers and terminals are present in close proximity to hindbrain catecholaminergic neurons, and fourth ventricular (4V) orexin injections that increase food intake also increase c-Fos expression in hindbrain catecholamine neurons, suggesting that orexin neurons may stimulate feeding by activating catecholamine neurons. Here we examine that hypothesis in more detail. We found that 4V injection of orexin-A (0.5 nmol/rat) produced widespread activation of c-Fos in hindbrain catecholamine cell groups. In the A1 and C1 cell groups in the ventrolateral medulla, where most c-Fos-positive neurons were also dopamine ß hydroxylase (DBH) positive, direct injections of a lower dose (67 pmol/200 nl) of orexin-A also increased food intake in intact rats. Then, with the use of the retrogradely transported immunotoxin, anti-DBH conjugated to saporin (DSAP), which targets and destroys DBH-expressing catecholamine neurons, we examined the hypothesis that catecholamine neurons are required for orexin-induced feeding. Rats given paraventricular hypothalamic injections of DSAP, or unconjugated saporin (SAP) as control, were implanted with 4V or lateral ventricular (LV) cannulas and tested for feeding in response to ventricular injection of orexin-A (0.5 nmol/rat). Both LV and 4V orexin-A stimulated feeding in SAP controls, but DSAP abolished these responses. These results reveal for the first time that catecholamine neurons are required for feeding induced by injection of orexin-A into either LV or 4V.


Subject(s)
Catecholamines/metabolism , Cerebral Ventricles/drug effects , Eating/drug effects , Feeding Behavior/drug effects , Intracellular Signaling Peptides and Proteins/administration & dosage , Nerve Fibers/drug effects , Neuropeptides/administration & dosage , Rhombencephalon/drug effects , Animals , Cerebral Ventricles/cytology , Cerebral Ventricles/immunology , Cerebral Ventricles/metabolism , Dopamine beta-Hydroxylase/immunology , Dopamine beta-Hydroxylase/metabolism , Immunotoxins/administration & dosage , Injections, Intraventricular , Male , Nerve Fibers/immunology , Nerve Fibers/metabolism , Orexins , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Rhombencephalon/cytology , Rhombencephalon/immunology , Rhombencephalon/metabolism , Ribosome Inactivating Proteins, Type 1/administration & dosage , Saporins
7.
Int J Med Sci ; 12(5): 397-406, 2015.
Article in English | MEDLINE | ID: mdl-26005374

ABSTRACT

The use of toxins for cancer therapy has great promise. Gelonin, a potent plant toxin, causes cell death by inactivating the 60S ribosomal subunit. Recently, we developed a novel gene delivery system using biodegradable cationic heparin-polyethyleneimine (HPEI) nanogels. In the current study, the antitumor activity of a recombinant plasmid expressing gelonin (pGelonin) on human ovarian cancer was assessed. The application of HPEI nanogels, was also evaluated. Gelonin-cDNA was cloned into the pVAX1 plasmid vector and transfected into SKOV3 human ovarian cancer cells using biodegradable cationic HPEI nanogels. The expression of gelonin in vitro and in vivo was confirmed using RT-PCR and western blot analysis. Cell viability and apoptosis were examined using an MTT assay and flow cytometric analysis. For the in vivo study, an SKOV3 intraperitoneal ovarian carcinomatosis model was established, and nude mice were randomly assigned into four groups receiving i.p. administration of pGelonin/HPEI complexes, pVAX/HPEI complexes, HPEI alone and 5% glucose solution. The tumor weight was monitored, and a TUNEL assay and Ki-67 immunohistochemistry were performed to evaluate apoptosis and cell proliferation in the tumor tissue sections, respectively. Gelonin was efficiently expressed in SKOV3 cancer cells in vitro and in vivo using pGelonin incorporated with HPEI nanogels. The pGelonin/HPEI complexes inhibited cell viability and induced apoptosis in the cell culture. Treatment for intraperitoneal carcinomatosis with pGelonin/HPEI complexes reduced the tumor weight by ~58.55% compared to the control groups (P<0.05). The antitumor effect was accompanied by increased apoptosis and reduced cell proliferation (P<0.05). No significant side effects were observed with i.p. administration of the pGelonin/HPEI complexes. Our data indicate that HPEI nanogel-delivered pGelonin may have promising applications against human ovarian cancer.


Subject(s)
Genetic Therapy/methods , Heparin/chemistry , Ovarian Neoplasms/therapy , Polyethylene Glycols/administration & dosage , Polyethyleneimine/administration & dosage , Ribosome Inactivating Proteins, Type 1/pharmacology , Animals , Apoptosis/genetics , Biocompatible Materials , Cations , Cell Line , Female , Gene Transfer Techniques , Heparin/administration & dosage , Humans , Mice, Inbred BALB C , Mice, Nude , Nanogels , Ovarian Neoplasms/pathology , Polyethylene Glycols/chemistry , Polyethylene Glycols/toxicity , Polyethyleneimine/chemistry , Polyethyleneimine/toxicity , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/genetics , Xenograft Model Antitumor Assays
8.
Immunopharmacol Immunotoxicol ; 37(1): 42-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25347443

ABSTRACT

CONTEXT: Saponinum album (SA) is a complex mixture of triterpenoid saponins previously shown to augment the cytotoxicity of the type I ribosome-inactivating protein saporin and an EGF-saporin target toxin that could potentially be used to improve the therapeutic window of targeted toxins. OBJECTIVE: To investigate the augmentative property of SA on saporin and saporin-based immunotoxins (IT) directed against five different cell surface target molecules on human leukemia and lymphoma cells. MATERIALS AND METHODS: After determining the optimum dose of SA for each cell line, the extent of SA-mediated augmentation was established for saporin and five saporin-based ITs using XTT and an annexin V apoptosis assay. Immunospecificity was investigated using three different blocking assays. Dose-scheduling was also investigated using the XTT assay. RESULTS: Uncorrected SA-mediated augmentation ranged at best from 31.5 million-fold to, at worse, 174-fold. However, when the calculated fold-increases were adjusted for the non-immunospecific effects of SA on an off-target IT, the true augmentative effects of SA were found to be largely non-immunospecific. Antibody blocking studies demonstrated that the augmentative effect of SA was only partially immunospecific. Separate exposure of target cells to IT and SA at different times demonstrated that immunospecific augmentation of IT by SA could be achieved but only if cells were exposed to IT first and SA second. CONCLUSIONS: SA significantly, although variably, augments the cytotoxicity of saporin and saporin-based immunotoxins. Concomitant exposure to both IT and SA can result in non-immunospecific cytotoxicity that can be overcome by temporally separating exposure to each.


Subject(s)
Apoptosis/drug effects , Cytotoxicity, Immunologic/drug effects , Ribosome Inactivating Proteins, Type 1/pharmacology , Saponins/pharmacology , Triterpenes/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/immunology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/immunology , Burkitt Lymphoma/immunology , Burkitt Lymphoma/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Administration Schedule , Flow Cytometry , Humans , Molecular Targeted Therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/isolation & purification , Saponins/administration & dosage , Saponins/isolation & purification , Saporins , Triterpenes/administration & dosage , Triterpenes/isolation & purification
9.
Georgian Med News ; (240): 59-64, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25879561

ABSTRACT

The aim of this study was to investigate the role of the medial septal (MS) GABAergic cells in hippocampal dependent spatial learning using the immunotoxin GAT1-SAP to produce selective lesions of GABAergic MS neurons. In current study rats were trained in a visible platform version of the Morris water maze in which either a place or cue strategy could be used to escape successfully. Immunohistochemical studies showed that intraseptal injection of GAT1-SAP extensively damaged GABAergic MS neurons and spared most cholinergic neurons. The rats' responses on the competition test were classified as either cue or place, based on the swim path for those trials. An overview of the data from both competition trials for each group show that the control rats in 14 trials out of 16 competition test trial used place strategy, while MS-lesioned ones used this strategy in 2 trials only. Decreased place-bias in MS-lesioned rats compared to the control rats was significant (P<0.01). The data obtained in the control and GAT1-SAP lesioned animals in the present study, demonstrate that lesioned rats were impaired in hidden platform trials during training, and displayed a pronounced cue-bias in competition tests. Therefore, above data suggest involvement of the MS GABAergic neurons in organization of the spatial map-driven behavior and this structure, along with the hippocampus, should be viewed as a constituent of the functional system responsible for the cognitive types of spatial memory.


Subject(s)
GABAergic Neurons/pathology , Ribosome Inactivating Proteins, Type 1/administration & dosage , Septal Nuclei/physiopathology , Spatial Learning/drug effects , Animals , GABAergic Neurons/drug effects , Humans , Immunotoxins/administration & dosage , Maze Learning/drug effects , Rats , Saporins , Septal Nuclei/drug effects , gamma-Aminobutyric Acid/metabolism
10.
Georgian Med News ; (239): 98-103, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25802458

ABSTRACT

In the present study electrolytic and the immunotoxins (192 IgG saporin and GAT1-SAP) lesions of medial septal area (MS) were used to investigate the importance of cholinergic and GABAergic MS neurons in spatial working memory using spatial alternation task. In our experiments electrolytic lesions destroyed on average 69% of the intact MS. Examination of the AChE stained sections showed that after injections of 192 IgG saporin into the MS, animals exhibited significantly less AChE staining in MS as compared to sections obtained from control animals. Intraseptal GAT1-SAP preferentially reduced GABAergic neurons as compared to cholinergic neurons in the MS. The results of present study indicate that spatial short-term memory is affected only by electrolytic but not 192 IgG saporin or GAT1-SAP lesions. The behavioral testing showed that 192 IgG saporin treated rats, relative to control rats, had a significantly lower level in the number of arms entered during the testing session. However, the groups did not differ in the level of alternation behavior. GAT1-SAP lesioned rats showed that the percent alternation scores and the number of arms that the rat entered in the maze were not significantly different from control rats. These findings indicate that deficits observed after septal electrolytic lesions cannot be accounted solely to the loss of cholinergic or GABAergic septohippocampal projections. To determine more definitively whether septohippocampal projection neurons are required for the spatial short-term memory it would be ideal to produce in future combined lesions of the cholinergic and GABA-ergic septohippocampal projection neurons using 192 IgG-saporin and GAT1-SAP.


Subject(s)
Memory, Short-Term/physiology , Neurons/physiology , Septum of Brain/physiology , Acetylcholine/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Hippocampus/drug effects , Hippocampus/physiology , Humans , Immunotoxins/administration & dosage , Male , Maze Learning/drug effects , Memory, Short-Term/drug effects , Neurons/drug effects , Rats , Ribosome Inactivating Proteins, Type 1/administration & dosage , Saporins , Septum of Brain/drug effects , gamma-Aminobutyric Acid/metabolism
11.
J Neurosci ; 33(15): 6659-71, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23575862

ABSTRACT

The nucleus basalis (NB) in the basal forebrain provides most of the cholinergic input to the neocortex and has been implicated in a variety of cognitive functions related to the processing of sensory stimuli. However, the role that cortical acetylcholine release plays in perception remains unclear. Here we show that selective loss of cholinergic NB neurons that project to the cortex reduces the accuracy with which ferrets localize brief sounds and prevents them from adaptively reweighting auditory localization cues in response to chronic occlusion of one ear. Cholinergic input to the cortex was disrupted by making bilateral injections of the immunotoxin ME20.4-SAP into the NB. This produced a substantial loss of both p75 neurotrophin receptor (p75(NTR))-positive and choline acetyltransferase-positive cells in this region and of acetylcholinesterase-positive fibers throughout the auditory cortex. These animals were significantly impaired in their ability to localize short broadband sounds (40-500 ms in duration) in the horizontal plane, with larger cholinergic cell lesions producing greater performance impairments. Although they localized longer sounds with normal accuracy, their response times were significantly longer than controls. Ferrets with cholinergic forebrain lesions were also less able to relearn to localize sound after plugging one ear. In contrast to controls, they exhibited little recovery of localization performance after behavioral training. Together, these results show that cortical cholinergic inputs contribute to the perception of sound source location under normal hearing conditions and play a critical role in allowing the auditory system to adapt to changes in the spatial cues available.


Subject(s)
Auditory Perception/physiology , Cerebral Cortex/physiology , Cholinergic Neurons/physiology , Neuronal Plasticity/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Auditory Cortex/physiology , Auditory Perception/drug effects , Basal Nucleus of Meynert/drug effects , Basal Nucleus of Meynert/physiology , Cell Death/drug effects , Cell Death/physiology , Cholinergic Neurons/drug effects , Ferrets , Immunotoxins/administration & dosage , Microinjections , Nerve Degeneration/chemically induced , Nerve Degeneration/psychology , Neural Pathways/drug effects , Neural Pathways/physiology , Neuronal Plasticity/drug effects , Recovery of Function/physiology , Ribosome Inactivating Proteins, Type 1/administration & dosage , Saporins , Sound Localization/drug effects , Sound Localization/physiology
12.
Biol Chem ; 395(12): 1461-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25205711

ABSTRACT

An immunotoxin composed of gelonin, a basic ribosome-inactivating protein, type I of 30 kDa, isolated from the seeds of the Indian plant Gelonium multiflorum and methotrexate (MTX) has been studied as a potential tool of gelonin delivery into the cytoplasm of MTX-responsive cells. On the average, about five molecules of methotrexate were chemically coupled to gelonin via an N-hydroxysuccinimide ester of the drug. The MTX-gelonin conjugate was able to reduce the viability of MCF-7 cells in a dose-dependent manner with ID50 of 10 nm, whereas gelonin or MTX alone showed none or very little effects. Besides its ribosome-inactivating activity, which is about ten-fold lower in an in vitro translation assay (IC50 of 50.5 ng/ml as compared to 4.6 ng/ml), the conjugate also significantly induced direct and oxidative DNA damage as shown by the alkaline comet assay. Hence, MTX-gelonin conjugates are promising candidates for the treatment of MTX-responsive cancers.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Immunotoxins/pharmacology , Methotrexate/pharmacology , Ribosome Inactivating Proteins, Type 1/pharmacology , Amino Acid Sequence , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Folate Receptors, GPI-Anchored/analysis , Folic Acid/analogs & derivatives , Folic Acid/analysis , Folic Acid/metabolism , Folic Acid Antagonists/administration & dosage , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Humans , Immunotoxins/administration & dosage , Immunotoxins/chemistry , MCF-7 Cells , Methotrexate/administration & dosage , Methotrexate/chemistry , Molecular Sequence Data , Protein Biosynthesis/drug effects , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/chemistry , Tetrahydrofolate Dehydrogenase/metabolism
13.
Biomater Sci ; 12(19): 5010-5022, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39177215

ABSTRACT

Saporin is a 28 621 Da protein and plant toxin possessing rRNA N-glycosidase activity. Due to its potent ribosome-inactivating ability, saporin is commonly studied as an anticancer agent. However, its enzymatic activity is greatly hindered by its poor plasma membrane permeability. To overcome this barrier, we used a bioinspired intracellular delivery platform based on the pH-responsive pseudopeptide, poly(L-lysine isophthalamide) grafted with L-phenylalanine at a stoichiometric molar percentage of 50% (PP50). PP50 was co-incubated with saporin (PP50/saporin) in a mildly acidic pH environment to aid intracellular delivery and increase saporin's therapeutic potential. We demonstrated that PP50 greatly enhanced the cytotoxicity of saporin in the 2D monolayer of A549 cells and 3D A549 multicellular spheroids whilst remaining non-toxic when administered alone. To elucidate the mechanism of cell death, we assessed the activation of caspases, the inhibition of protein synthesis, the onset of apoptosis and the mechanism of PP50/saporin entry. Inhibition of protein synthesis and activation of caspases 3/7, 8 and 9 were found to occur before the onset of apoptosis and cell death. PP50/saporin was also shown to rely on micropinocytosis and caveolae-mediated endocytosis for cell entry. In addition, fluorescein isothiocyanate-labelled saporin (FITC-saporin) was localized within the cytoplasm and nuclei when delivered with Cyanine5-labelled PP50 (Cy5-PP50). Taken together, this suggests that multiple pathways are triggered to initiate apoptosis and cell death in cells treated with PP50/saporin. Therefore, these results make PP50 a potential intracellular delivery platform for the internalization of protein therapeutics.


Subject(s)
Apoptosis , Saporins , Humans , Saporins/chemistry , Saporins/pharmacology , Apoptosis/drug effects , A549 Cells , Polylysine/chemistry , Polylysine/pharmacology , Caspases/metabolism , Hydrogen-Ion Concentration , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Ribosome Inactivating Proteins/chemistry , Ribosome Inactivating Proteins/pharmacology , Drug Delivery Systems , Peptides/chemistry , Peptides/pharmacology , Ribosome Inactivating Proteins, Type 1/pharmacology , Ribosome Inactivating Proteins, Type 1/chemistry , Ribosome Inactivating Proteins, Type 1/administration & dosage , Cell Survival/drug effects
14.
Biomacromolecules ; 14(4): 1093-102, 2013 Apr 08.
Article in English | MEDLINE | ID: mdl-23444913

ABSTRACT

Plant-derived Type I toxins are candidate anticancer therapeutics requiring cytosolic delivery into tumor cells. We tested a concept for two-stage delivery, whereby tumor cells precoated with an antibody-targeted gelonin toxin were killed by exposure to endosome-disrupting polymer nanoparticles. Co-internalization of particles and tumor cell-bound gelonin led to cytosolic delivery and >50-fold enhancement of toxin efficacy. This approach allows the extreme potency of gelonin to be focused on tumors with significantly reduced potential for off-target toxicity.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents/administration & dosage , Drug Carriers , Endosomes/metabolism , Neoplasms/drug therapy , Phalloidine/administration & dosage , Ribosome Inactivating Proteins, Type 1/administration & dosage , Animals , Antibodies , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Dextrans/metabolism , Humans , Mice , Nanoparticles , Ribosome Inactivating Proteins, Type 1/chemistry , Ribosome Inactivating Proteins, Type 1/pharmacology
15.
J Sleep Res ; 22(6): 721-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23701447

ABSTRACT

Cholinergic basal forebrain structures are implicated in cortical arousal and regulation of the sleep-wake cycle. Cholinergic neurones are innervated by noradrenergic terminals, noradrenaline excites them via alpha-1 receptors and microinjection of noradrenaline into the basal forebrain enhances wakefulness. However, it is not known to what extent the cholinergic versus non-cholinergic basal forebrain projection neurones contribute to the arousing effects of noradrenaline. To elucidate the roles of cholinergic basal forebrain structures we administered methoxamine, an alpha-1-adrenergic agonist into the basal forebrain, in intact animals and again after selective destruction of the basal forebrain cholinergic cells by 192 IgG-saporin. In eight male Han-Wistar rats implanted with electroencephalogram/electromyogram electrodes, a microdialysis probe targeted into the basal forebrain was perfused with artificial cerebrospinal fluid for 6 h on a baseline day, and with cerebrospinal fluid in the first and with methoxamine in the second 3-h period of the subsequent day. The sleep-wake activity was recorded for 24 h on both days. Saporin was then injected into the basal forebrain and 2 weeks later the same experimental schedule (with cerebrospinal fluid and methoxamine) was repeated. In the intact animals, methoxamine exhibited a robust arousing effect and non-rapid eye movement (NREM) and REM sleep was suppressed. Lesioning of the basal forebrain cholinergic neurones abolished almost completely the NREM sleep-suppressing effect of methoxamine, whereas the REM sleep-suppressing effect remained intact. Thus, the basal forebrain cholinergic neurones mediate, at least in part, cortical arousal and non-REM sleep-suppression, but they are not involved in the REM sleep-suppressing effects of noradrenaline.


Subject(s)
Arousal/drug effects , Arousal/physiology , Cholinergic Neurons/drug effects , Norepinephrine/pharmacology , Prosencephalon/drug effects , Prosencephalon/physiology , Sleep Stages/drug effects , Animals , Antibodies, Monoclonal , Cholinergic Neurons/physiology , Electroencephalography/drug effects , Male , Methoxamine/administration & dosage , Methoxamine/pharmacology , Microdialysis , Prosencephalon/anatomy & histology , Prosencephalon/cytology , Rats , Rats, Wistar , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins , Sleep Stages/physiology , Wakefulness/drug effects , Wakefulness/physiology
16.
Neoplasma ; 60(5): 469-79, 2013.
Article in English | MEDLINE | ID: mdl-23790164

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is one of the most common types of cancer worldwide. However, there is currently no effective therapy strategy in the clinical practice. Recombinant phytotoxin gelonin fused to other factors have been used to treat different cancers. But there have been no reports of gelonin gene therapy. In this study, we have constructed a recombinant plasmid which contained a tumor-specific survivin promoter to drive phytotoxin gelonin (pSur-Gel). And the cytotoxicity effects of pSur-Gel in HCC were also validated both in vitro and in vivo. The expression level of survivin was detected in different liver cancer cell lines and normal liver cell lines by western blot analysis, and a survivin promoter-driven green fluorescent protein (GFP) expression vectors (pSur-GFP) was also tested in liver cancer cell line HepG2 and normal liver cell line LO2. Moreover, phytotoxin gelonin expression experiment and cytotoxicity experiment of pSur-Gel was performed in HepG2 cells and LO2 cells in vitro. Furthermore, anti-tumor effect of pSur-Gel against HepG2 xenografts and toxicity of this gene were evaluated in the mice model. Finally, LDH release assay, apoptosis assay and immunoblot analyse LC3 conversion (LC3-I to LC3-II) were tested. We found that the expression of survivin protein was higher in liver cancer cell lines compared with the normal liver cells. Further study showed that the pSur-GFP and pSur-Gel was expressed specially in liver cancer cell other than in normal liver cells. pSur-Gel plasmid could effectively inhibit the proliferation of liver cancer cells (*P<0.05), and significantly repress the growth of HepG2 xenografts via intravenous in vivo (*P<0.05). Otherwise, compared to cytomegalovirus promoter-driven gelonin expression vectors (pCMV-Gel), no significantly systemic toxicity or organ injuries had been observed in pSur-Gel treated mice. Further studies revealed that the phytotoxin gelonin induced cell death might be mediated by apoptosis and the damage of cell membrane. Taken together, treating hepatocellular carcinoma with the pSur-Gel may be a novel and interesting cancer gene therapy protocol and is worthy of further development for future clinical trials. KEYWORDS: liver cancer, gelonin, survivin promoter, gene therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Carcinoma, Hepatocellular/therapy , Genetic Therapy/methods , Inhibitor of Apoptosis Proteins/genetics , Liver Neoplasms/therapy , Ribosome Inactivating Proteins, Type 1/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Ribosome Inactivating Proteins, Type 1/adverse effects , Survivin , Transfection , Xenograft Model Antitumor Assays
17.
Eur J Orthop Surg Traumatol ; 23(3): 263-72, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23412299

ABSTRACT

We employed 54 rats to devise a model of neuroma-in-continuity and explore the effect of the immunotoxin OX7-saporin on the neuroma. The left common peroneal, tibial or sciatic nerves were crushed by one 10-s application of a micro-artery forceps. At 3 and 6 weeks, the nerve was cut distal to the site of nerve crush, and retrograde fluorescent labeling was done. Pressure microinjection of 2 µl of natural saline or 2 µl of the immunotoxin conjugate OX7-saporin was done at the nerve stump 2 days later. Sacrifice was done after 3 weeks. In all control and saline-injection nerve specimens, gross observation and histology showed a neuroma-in-continuity. In 19 of the 24 OX7-saporin nerve specimens, gross observation showed a narrowed area at the site of nerve crush. Histology showed inhibition of neuroma-in-continuity formation. Fluorescent microscopy showed ablation of the labeled neurons in the dorsal root ganglia corresponding to the OX7-saporin subgroups.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunoconjugates/therapeutic use , Immunotoxins/therapeutic use , Peripheral Nerve Injuries/drug therapy , Ribosome Inactivating Proteins, Type 1/therapeutic use , Animals , Antibodies, Monoclonal/administration & dosage , Disease Models, Animal , Ganglia, Spinal/drug effects , Immunoconjugates/administration & dosage , Immunotoxins/administration & dosage , Male , Microinjections/methods , Microscopy, Fluorescence , Nerve Crush , Peripheral Nerve Injuries/pathology , Peroneal Nerve/injuries , Peroneal Nerve/pathology , Rats , Ribosome Inactivating Proteins, Type 1/administration & dosage , Saporins , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Tibial Nerve/injuries , Tibial Nerve/pathology
18.
Hippocampus ; 22(4): 914-25, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21542057

ABSTRACT

The vestibular system has been suggested to participate in spatial navigation, a function ascribed to the hippocampus. Vestibular stimulation during spatial navigation activates a hippocampal theta rhythm (4-10 Hz), which may enhance spatial processing and motor response. We hypothesize that a cholinergic, atropine-sensitive theta is generated during passive whole-body rotation in freely behaving rats. Hippocampal EEGs were recorded by implanted electrodes in CA1 while rats were rotated on a vertical axis, for a minute or longer, at different angular velocities. Rotation induced a continuous hippocampal theta rhythm while the rat was immobile, in both light and dark conditions. Theta peak frequency showed a significant increase during high (50-70 rpm) as compared with a lower (20-49 rpm) rotational velocity. Rotation-induced theta was abolished by muscarinic receptor antagonist atropine sulfate (50 mg/kg i.p.) but not by atropine methyl nitrate (50 mg/kg i.p.), which did not pass the blood-brain barrier. Theta was attenuated in rats in which cholinergic neurons in the medial septum (MS) were lesioned with 192 IgG-saporin (0.14 µg in 0.4 µl), as confirmed by depletion of MS cells immunoreactive to choline acetyltransferase and an absence of acetylcholinesterase staining in the hippocampus. Bilateral lesion of the vestibular receptors by sodium arsanilate (30 mg in 0.1 ml, intratympanically) also attenuated the rotation-induced theta rhythm. In intact rats, field excitatory postsynaptic potentials (fEPSPs) in CA1 evoked by commissural stimulation were smaller during walking or rotation as compared with during immobility. Modulation of fEPSP was absent following atropine sulfate in intact rats and in 192 IgG-saporin lesion rats. In summary, this is the first report of a continuous atropine-sensitive hippocampal theta in the rat induced by vestibular stimulation during rotation, and accompanied by cholinergic modulation of hippocampal synaptic transmission. Vestibular-activated septohippocampal cholinergic activity could be an important component in sensorimotor processing and spatial memory.


Subject(s)
Hippocampus/physiology , Theta Rhythm/physiology , Vestibule, Labyrinth/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Atropine/administration & dosage , Atropine Derivatives/administration & dosage , Blood-Brain Barrier , Choline O-Acetyltransferase/metabolism , Cholinergic Neurons/drug effects , Cholinergic Neurons/physiology , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Hippocampus/injuries , Immobilization/physiology , Immunotoxins/administration & dosage , Male , Muscarinic Antagonists/administration & dosage , Rats , Rats, Long-Evans , Ribosome Inactivating Proteins, Type 1/administration & dosage , Rotation , Saporins , Theta Rhythm/drug effects , Walking/physiology
19.
Mol Pain ; 8: 35, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22540287

ABSTRACT

BACKGROUND: A small proportion of lamina I neurons of the spinal cord project upon the hindbrain and are thought to engage descending pathways that modulate the behavioural response to peripheral injury. Early postnatal development of nociception in rats is associated with exaggerated and diffuse cutaneous reflexes with a gradual refinement of responses over the first postnatal weeks related to increased participation of inhibitory networks. This study examined the postnatal development of lamina I projection neurons from postnatal day 3 (P3) until P48. RESULTS: At P3, a subset of lamina I neurons were found to express the neurokinin 1 (NK1) receptor. Using fluorogold retrograde tracing, we found that the NK1 positive neurons projected upon the parabrachial nucleus (PB) within the hindbrain. Using c-fos immunohistochemistry, we showed that lamina I and PB neurons in P3 rats responded to noxious stimulation of the periphery. Finally, ablation of lamina I neurons with substance-P saporin conjugates at P3 resulted in increased mechanical sensitivity from P45 onwards compared to control animals of the same age. CONCLUSIONS: These results suggest that the lamina I pathway is present and functional at least from P3 and required for establishing and fine-tuning mechanical sensitivity in adult rats.


Subject(s)
Aging/metabolism , Mechanotransduction, Cellular , Neurons/metabolism , Receptors, Neurokinin-1/metabolism , Sensory Thresholds , Animals , Animals, Newborn , Male , Mechanotransduction, Cellular/drug effects , Neurons/drug effects , Physical Stimulation , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Posterior Horn Cells/pathology , Posterior Horn Cells/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins , Sensory Thresholds/drug effects , Substance P/administration & dosage , Substance P/analogs & derivatives , Substance P/pharmacology , Weight Gain/drug effects
20.
J Immunol ; 184(8): 4196-204, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20220085

ABSTRACT

There is compelling evidence that self-reactive CD8(+) T cells are a major factor in development and progression of type 1 diabetes in animals and humans. Hence, great effort has been expended to define the specificity of autoimmune CD8(+) T cells and to alter their responses. Much work has focused on tolerization of T cells using proteins or peptides. A weakness in this approach is that residual autoreactive T cells may be activated and exacerbate disease. In this report, we use a novel approach, toxin-coupled MHC class I tetramers. Used for some time to identify Ag-specific cells, in this study, we use that same property to delete the Ag-specific cells. We show that saporin-coupled tetramers can delete islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-reactive T cells in vitro and in vivo. Sequence analysis of TCRbeta-chains of IGRP(+) cells reveals the repertoire complexity in the islets is markedly decreased as NOD mice age and significantly altered in toxic tetramer-treated NOD mice. Further tetramer(+) T cells in the islets are almost completely deleted, and, surprisingly, loss of tetramer(+) T cells in the islets is long lasting. Finally, we show deletion at 8 wk of age of IGRP(+) CD8(+) T cells, but not dystophia myotonica kinase- or insulin B-reactive cells, significantly delays diabetes in NOD mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Diabetes Mellitus, Type 1/prevention & control , H-2 Antigens/administration & dosage , Immunotoxins/administration & dosage , Ribosome Inactivating Proteins, Type 1/toxicity , beta 2-Microglobulin/administration & dosage , Animals , Autoantigens/immunology , Autoantigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Death/immunology , Cell Movement/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Disease Progression , Epitopes, T-Lymphocyte/immunology , Female , Glucose-6-Phosphatase/administration & dosage , Glucose-6-Phosphatase/biosynthesis , Glucose-6-Phosphatase/immunology , H-2 Antigens/toxicity , Histocompatibility Antigen H-2D , Immunotoxins/toxicity , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Molecular Mimicry/immunology , Proteins/administration & dosage , Proteins/immunology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Ribosome Inactivating Proteins, Type 1/administration & dosage , Saporins , beta 2-Microglobulin/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL