Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1331345, 2024.
Article in English | MEDLINE | ID: mdl-38370401

ABSTRACT

Chimeric antigen receptor (CAR) T cell technology has ushered in a new era of immunotherapy, enabling the targeting of a broad range of surface antigens, surpassing the limitations of traditional T cell epitopes. Despite the wide range of non-protein tumor-associated antigens, the advancement in crafting CAR T cells for these targets has been limited. Owing to an evolutionary defect in the CMP-Neu5Ac hydroxylase (CMAH) that abolishes the synthesis of CMP-Neu5Gc from CMP-Neu5Ac, Neu5Gc is generally absent in human tissues. Despite this, Neu5Gc-containing antigens, including the ganglioside GM3(Neu5Gc) have consistently been observed on tumor cells across a variety of human malignancies. This restricted expression makes GM3(Neu5Gc) an appealing and highly specific target for immunotherapy. In this study, we designed and evaluated 14F7-28z CAR T cells, with a targeting unit derived from the GM3(Neu5Gc)-specific murine antibody 14F7. These cells exhibited exceptional specificity, proficiently targeting GM3(Neu5Gc)-expressing murine tumor cells in syngeneic mouse models, ranging from B cell malignancies to epithelial tumors, without compromising safety. Notably, human tumor cells enhanced with murine Cmah were effectively targeted and eliminated by the 14F7 CAR T cells. Nonetheless, despite the detectable presence of GM3(Neu5Gc) in unmodified human tumor xenografts, the levels were insufficient to trigger a tumoricidal T-cell response with the current CAR T cell configuration. Overall, our findings highlight the potential of targeting the GM3(Neu5Gc) ganglioside using CAR T cells across a variety of cancers and set the stage for the optimization of 14F7-based therapies for future human clinical application.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Animals , Mice , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/therapeutic use , G(M3) Ganglioside/therapeutic use , Antigens, Neoplasm
2.
J Med Chem ; 65(3): 1883-1897, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35073068

ABSTRACT

Cancer is a leading cause of death worldwide. Recent research studies have revealed that GM3 derivatives have considerable promise as potential therapeutic agents for cancer. To discover novel GM3 derivatives as potential antitumor agents, a one-pot enzymatic synthesis was established, yielding 14 GM3 derivatives in high total yields (22-41%). Subsequently, the inhibitory activities of GM3 derivatives were assessed by wound-healing assays and Transwell assays and tumor-bearing animal models. Among all the GM3 derivatives, N-12 showed excellent migration and invasion inhibitory effects in cells and marked antitumor activity in C57BL/6 mice. The subsequent analysis of cancer tissues and serum samples revealed that N-12 induces tumor inhibition, which was closely related to immune response. Taken together, N-12 can be further developed as an effective therapeutic for the treatment of cancer. An RNA-sequencing (RNA-seq) analysis was then performed and indicated that the antitumor mechanism of N-12 involved focal adhesion and ECM-receptor interaction signaling pathways.


Subject(s)
Antineoplastic Agents/therapeutic use , G(M3) Ganglioside/analogs & derivatives , G(M3) Ganglioside/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cytokines/metabolism , G(M3) Ganglioside/chemical synthesis , G(M3) Ganglioside/pharmacokinetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunity/drug effects , Immunotherapy , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Necrosis/chemically induced , Signal Transduction/drug effects
3.
Int J Mol Sci ; 22(24)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34948420

ABSTRACT

Recently, the atheroprotective role of endogenous GM3 and an atherogenesis-inhibiting effect of exogenous GM3 suggested a possibility of exogenous GM3 being recruited as an anti-atherosclerotic drug. This study seeks to endow exogenous GM3 with atherosclerotic targetability via reconstituted high-density lipoprotein (rHDL), an atherosclerotic targeting drug nanocarrier. Unloaded rHDL, rHDL loaded with exogenous GM3 at a low concentration (GM3L-rHDL), and rHDL carrying GM3 at a relatively high concentration (GM3H-rHDL) were prepared and characterized. The inhibitory effect of GM3-rHDL on lipid deposition in macrophages was confirmed, and GM3-rHDL did not affect the survival of red blood cells. In vivo experiments using ApoE-/- mice fed a high fat diet further confirmed the anti-atherosclerotic efficacy of exogenous GM3 and demonstrated that GM3 packed in HDL nanoparticles (GM3-rHDL) has an enhanced anti-atherosclerotic efficacy and a reduced effective dose of GM3. Then, the macrophage- and atherosclerotic plaque-targeting abilities of GM3-rHD, most likely via the interaction of ApoA-I on GM3-rHDL with its receptors (e.g., SR-B1) on cells, were certified via a microsphere-based method and an aortic fragment-based method, respectively. Moreover, we found that solution acidification enhanced GM3 release from GM3-rHDL nanoparticles, implying the pH-responsive GM3 release when GM3-rHDL enters the acidic atherosclerotic plaques from the neutral blood. The rHDL-mediated atherosclerotic targetability and pH-responsive GM3 release of GM3-rHDL enhanced the anti-atherosclerotic efficacy of exogenous GM3. The development of the GM3-rHDL nanoparticle may help with the application of exogenous GM3 as a clinical drug. Moreover, the data imply that the GM3-rHDL nanoparticle has the potential of being recruited as a drug nanocarrier with atherosclerotic targetability and enhanced anti-atherosclerotic efficacy.


Subject(s)
Atherosclerosis/drug therapy , G(M3) Ganglioside/pharmacology , Lipoproteins, HDL , Macrophages/drug effects , Nanoparticles/chemistry , Plaque, Atherosclerotic/drug therapy , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Drug Delivery Systems , G(M3) Ganglioside/therapeutic use , Humans , Hydrogen-Ion Concentration , Lipid Metabolism , Macrophages/metabolism , Male , Mice , Mice, Knockout, ApoE , RAW 264.7 Cells
4.
Pak J Pharm Sci ; 28(2 Suppl): 701-4, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25796161

ABSTRACT

This paper aims to make an analysis of the effects of ganglioside (GM) combined with methylprednisolone (MP) in early acute spinal injury. Fifty-three patients with acute spinal cord injury were included in this study and they were randomly divided into experimental and control group. Twenty-seven patients in the control group were treated with MP, while the rest 26 patients received more GM based on that. By observing and comparing the clinical responses from patients and recovery time of all indexes, results came out: the curative rates in the experimental and control group were 50.0%, 40.7% respectively, and the total effective rates were 92.3%, 85.2% respectively. There was a remarkable difference between the two groups (P<0.05). Patients in the experimental group took 6.2 ± 1.9d to restore their sphincter function, 11.2 ± 2.8d to recover their muscle forces to over grade II, and 13.8 ± 3.9d to return general activity, while the patients in the other group clearly spent longer time on recovery, that were 12.1 ± 3.2, 19.2 ± 4.6 and 23.9 ± 5.6 respectively. The distinct difference between the two groups was of statistical significance (P<0.05). We conclude that GM has better curative effects than MP, for it is able to promote the recovery of nerve function for patients and greatly improve the prognosis.


Subject(s)
G(M3) Ganglioside/therapeutic use , Glucocorticoids/therapeutic use , Methylprednisolone/therapeutic use , Spinal Injuries/drug therapy , Acute Disease , Adolescent , Adult , China , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Neurologic Examination , Recovery of Function , Spinal Injuries/diagnosis , Spinal Injuries/physiopathology , Time Factors , Treatment Outcome , Young Adult
5.
Urology ; 81(1): 210.e11-5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23102779

ABSTRACT

OBJECTIVE: To investigate the therapeutic effects of exogenous gangliosides GM3 on human bladder cancer cell lines and the severe combined immunodeficiency mouse model of orthotopic bladder cancer. MATERIALS AND METHODS: Human bladder cancer cell lines YTS-1, T24, 5637, and KK47 were used in the study. In vitro cytotoxicity of GM3 was assessed using the cell counting kit-8. Cell adhesion was determined using a spreading assay. Phosphorylation of epidermal growth factor receptor was determined by Western blotting. In vivo, the orthotopic bladder cancer model was established using severe combined immunodeficiency mice and GM3 was administered intravesically by way of a transurethral catheter. RESULTS: GM3 inhibited the proliferation of all the bladder cancer cell lines tested. The addition of GM3 decreased cell adhesion and epidermal growth factor-dependent phosphorylation of epidermal growth factor receptor. Direct instillation of GM3 into the bladder of the orthotopic model significantly inhibited tumor growth. CONCLUSION: Our results suggest exogenous GM3 as a potential therapeutic agent for treating bladder cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , G(M3) Ganglioside/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Administration, Intravesical , Animals , Antineoplastic Agents/administration & dosage , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , ErbB Receptors/metabolism , Female , G(M3) Ganglioside/administration & dosage , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Phosphorylation/drug effects , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
6.
Biochim Biophys Acta ; 1788(12): 2518-25, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19837046

ABSTRACT

Therapeutic vaccination with tumor antigens is a new approach in cancer treatment, which aims at inducing immune response while avoiding the side effects generally associated to many conventional therapies. To improve the efficacy of vaccines, suitable carriers may be used. Herein the insertion of a thioether analogue of GM3 lactone (SNeuAC-C14) into liposomes is reported. SNeuAC-C14 is a potential vaccine for the targeting of saccharide-based tumor epitopes. Different liposome formulations were designed to act as carriers and to generate recognition by tumor epitopes. The structural study of pure and loaded liposomes was carried out by synchrotron Small Angle X-ray Scattering and was complemented by Dynamic Light Scattering and Zeta potential measurements. This provided detailed information on relevant properties of the investigated host-guest structures and showed that the active unit of SNeuAC-C14, i.e. its spiro tricyclic moiety, was located in the polar head region of the liposome bilayer, which is an important requirement for recognition phenomena. Moreover, it was found that most of the SNeuAC-C14/liposome complexes were positively charged. The obtained results allow these systems to be considered as candidates to promote immunoresponse in tumor cells.


Subject(s)
Cancer Vaccines/chemistry , Epitopes/chemistry , G(M3) Ganglioside/analogs & derivatives , Lipid Bilayers , Cancer Vaccines/therapeutic use , Epitopes/therapeutic use , G(M3) Ganglioside/chemistry , G(M3) Ganglioside/therapeutic use , Liposomes/chemistry , Liposomes/therapeutic use , Neoplasms/therapy , Structure-Activity Relationship
7.
Cancer Biol Ther ; 6(6): 853-5, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17611391

ABSTRACT

A case of a 16-year-old female with a disseminated tumor was reported six years after surgical treatment. In October 1993 the diagnosis of a bronchial carcinoid tumor was made and a left pneumonectomy was performed. No adjuvant treatment was indicated. In May 1999 a relapse was confirmed by cytology, and a treatment with rec-hIFNalpha (10 MU intramuscular, thrice/week) was indicated. Her clinical condition improved, (ECOG 2 to 0), after three months of stable disease at evaluation, up to March 2002 when she developed a progressive disease as documented by abdominal CT scan. The histological block of the primary tumor was sent to the Centre of Molecular Immunology in order to evaluate the recognition of the ganglioside molecule on the tumor by immunohistochemistry, which was informed as positive. In April 2002 we decided to begin a compassionate treatment with the vaccine NGcGM3/VSSP Montanide ISA 51 in combination with rec-hIFNalpha. Since then up to now (56 months after progression with rec-hIFNalpha alone) the patient still has stable disease. In summary, we observed very encouraging results that could support further studies in this type of patients.


Subject(s)
Bronchial Neoplasms/therapy , Cancer Vaccines/therapeutic use , Carcinoid Tumor/therapy , G(M3) Ganglioside/analogs & derivatives , Interferon-alpha/therapeutic use , Interferons/therapeutic use , Adolescent , Disease Progression , Disease-Free Survival , Female , G(M3) Ganglioside/immunology , G(M3) Ganglioside/therapeutic use , Humans , Immunohistochemistry/methods , Recurrence , Remission Induction , Tomography, X-Ray Computed
8.
Glycobiology ; 16(7): 573-83, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16574813

ABSTRACT

The simple ganglioside GM3 has been shown to have anti-proliferative effects in several in vitro and in vivo cancer models. Although the exogenous ganglioside GM3 has an inhibitory effect on cancer cell proliferation, the exact mechanism by which it prevents cell proliferation remains unclear. Previous studies showed that MDM2 is an oncoprotein that controls tumorigenesis through both p53-dependent and p53-independent mechanisms, and tumor suppressor phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a dual-specificity phosphatase that antagonizes phosphatidylinositol 3-kinase (PI-3K)/AKT signaling, is capable of blocking MDM2 nuclear translocation and destabilizing the MDM2 protein. Results from our current study show that GM3 treatment dramatically increases cyclin-dependent kinase (CDK) inhibitor (CKI) p21(WAF1) expression through the accumulation of p53 protein by the PTEN-mediated inhibition of the PI-3K/AKT/MDM2 survival signaling in HCT116 colon cancer cells. Moreover, the data herein clearly show that ganglioside GM3 induces p53-dependent transcriptional activity of p21(WAF1), as evidenced by the p21(WAF1) promoter-driven luciferase reporter plasmid (full-length p21(WAF1) promoter and a construct lacking the p53-binding sites). Additionally, ganglioside GM3 enhances expression of CKI p27(kip1) through the PTEN-mediated inhibition of the PI-3K/AKT signaling. Furthermore, the down-regulation of the cyclin E and CDK2 was clearly observed in GM3-treated HCT116 cells, but the down-regulation of cyclin D1 and CDK4 was not. On the contrary, suppression of PTEN levels by RNA interference restores the enhanced expression of p53-dependent p21(WAF1) and p53-independent p27(kip1) through inactivating the effect of PTEN on PI-3K/AKT signaling modulated by ganglioside GM3. These results suggest that ganglioside GM3-stimulated PTEN expression modulates cell cycle regulatory proteins, thus inhibiting cell growth. We conclude that ganglioside GM3 represents a modulator of cancer cell proliferation and may have potential for use in colorectal cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/genetics , G(M3) Ganglioside/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , PTEN Phosphohydrolase/metabolism , Antineoplastic Agents/analysis , Antineoplastic Agents/therapeutic use , Cell Cycle/drug effects , Cell Line, Tumor , Cell Membrane/chemistry , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Cyclin-Dependent Kinase Inhibitor p27 , Down-Regulation , G(M3) Ganglioside/analysis , G(M3) Ganglioside/therapeutic use , Humans , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
9.
J Clin Oncol ; 21(6): 1015-21, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12637465

ABSTRACT

PURPOSE: A heterophilic ganglioside cancer vaccine was developed by combining NeuGcGM3 with the outer membrane protein complex of Neisseria meningitidis to form very small size proteoliposomes (VSSP). A phase I clinical trial was performed to determine safety and immunogenicity of this vaccine. PATIENTS AND METHODS: Stage III to IV breast cancer patients received up to 15 (200 micro g) doses of the vaccine by intramuscular injection. The first five doses (induction phase) were given at 2-week intervals, with the remaining treatment (maintenance) administered on a monthly basis. RESULTS: Twenty-one patients, 11 of whom had metastatic disease, were included. Main toxicities included erythema and induration at the injection site, sometimes associated with mild pain, and low-grade fever (World Health Organization grades 1 and 2). All treated patients who completed the induction phase developed anti-NeuGcGM3 antibody titers between 1:1,280 and 1:164,000 immunoglobulin G (IgG), and 1:640 and 1:164,000 IgM. Noteworthy specific IgA antibodies were induced by vaccination in all stage III patients and in three stage IV patients. Serum antibody levels were higher in the stage III patients, with the larger increases observed after week 32. The antiganglioside IgG subclasses were mainly IgG1 and IgG3. Hyperimmune sera increased complement-mediated cytotoxicity versus P3X63 myeloma cells and a marked IgG differential reactivity against human mammary ductal carcinoma samples. CONCLUSION: NeuGcGM3/VSSP/Montanide ISA 51 is an unusual immunogenic ganglioside vaccine and also seems to be safe in this small trial. Immunologic surrogates of activity indicate that this reagent warrants further investigation.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Cancer Vaccines/therapeutic use , G(M3) Ganglioside/analogs & derivatives , G(M3) Ganglioside/therapeutic use , Adult , Aged , Breast Neoplasms/pathology , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Complement System Proteins/immunology , Enzyme-Linked Immunosorbent Assay , Female , G(M3) Ganglioside/administration & dosage , G(M3) Ganglioside/adverse effects , G(M3) Ganglioside/immunology , Humans , Immunoenzyme Techniques , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Immunohistochemistry , Injections, Intramuscular , Middle Aged , Neisseria meningitidis , Neoplasm Staging , Proteolipids , Remission Induction , Treatment Outcome
10.
Exp Neurol ; 168(2): 300-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259118

ABSTRACT

The simple ganglioside GM3 inhibits proliferation and induces apoptosis in proliferating immature rodent CNS cells. To determine whether GM3 influenced the expansion of human neural tumors the effects of GM3 treatment on primary human brain tumors were assayed. Here we demonstrate that GM3 treatment dramatically reduces cell numbers in primary cultures of high-grade human glioblastoma multiforme (GBM) tumors and the rat 9L cell gliosarcoma cell line. By contrast, GM3 treatment had little effect on cell number in cultures of normal human brain. A single injection of GM3 3 days after intracranial implantation of 9L tumor cells in a murine xenograft model system resulted in a significant increase in the symptom-free survival period of host animals. The effects of GM3 were not restricted to GBMs and 9L cells. Cultures of high-grade ependymomas, mixed gliomas, astrocytomas, oligodendrogliomas, and gangliogliomas were all susceptible to GM3 treatment. These results suggest that GM3 may have considerable value as a selectively toxic chemotherapeutic agent for human high-grade gliomas.


Subject(s)
Brain Neoplasms/drug therapy , G(M3) Ganglioside/therapeutic use , Glioma/drug therapy , Animals , Brain Neoplasms/pathology , Cell Death/drug effects , Cell Death/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Survival/drug effects , Cell Survival/physiology , Child, Preschool , Drug Screening Assays, Antitumor , G(M3) Ganglioside/pharmacology , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioma/pathology , Humans , Male , Mice , Rats , Transplantation, Heterologous , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/physiology
11.
Life Sci ; 57(13): PL165-9, 1995.
Article in English | MEDLINE | ID: mdl-7545772

ABSTRACT

Influence of FK-506 and ganglioside (GM3) on acquired immunity to Hymenolepis nana reinfection was examined in BALB/c mice. Treatment of mice with FK-506 abolished the acquired immunity to challenge infection with eggs of H. nana when the agent was injected inraperitoneally at daily doses of 9.0 to 11.0 mg/kg (but not 8.5 mg/kg) on days -1, 0, 1, 2, and 3 relative to the challenge. Intravenous injection of GM3 at a daily dose of 10.0 mg/kg could not produce an immunosuppressive effect on the acquired immunity. Combination treatment with 8.5 mg/kg/day FK-506 and 10.0 mg/kg/day of GM3 inhibited the acquired immunity. These results suggest that GM3 will be a good candidate for a clinical supplementary immunosuppressive agent.


Subject(s)
G(M3) Ganglioside/therapeutic use , Hymenolepiasis/therapy , Immunosuppressive Agents/therapeutic use , Tacrolimus/therapeutic use , Animals , Drug Synergism , Drug Therapy, Combination , Female , Hymenolepiasis/immunology , Immunity, Cellular , Mice , Mice, Inbred BALB C
15.
Exp Brain Res ; 60(1): 27-37, 1985.
Article in English | MEDLINE | ID: mdl-3930279

ABSTRACT

The effects of monosialoganglioside (GM1) injections on neuronal reorganization and behavioral recovery were studied in rats with unilateral transections of the nigro-striatal pathway. In Experiment 1, animals were treated daily with injections of saline or GM1 for not more than 14 days. At 2 days after surgery, GM1-treated animals exhibited less amphetamine-induced rotational asymmetry than did saline treated counterparts. This difference was still apparent at day 12, but vanished at postoperative day 39. Apomorphine-induced rotational asymmetry was equal in both groups at day 15, but by day 42, asymmetries increased in saline controls while remaining unchanged in GM1-treated animals. Rats were killed at either post-operative days 3, 15, or 45 after having received injections of horseradish peroxidase (HRP) into the denervated caudate nucleus. The number of neurons labelled by retrograde HRP-transport were counted in the ipsilateral substantia nigra pars compacta (iSNc), ipsilateral ventral tegmental area (iVTA), frontal cortex, and in the contralateral substantia nigra pars compacta (cSNc). Anterograde transport was also examined in the ipsilateral substantia nigra pars reticulata (iSNr). A significant loss of retrograde labelling in iSNc and iVTA was observed for both groups at post-operative day 3. At day 15, however, GM1-treated animals showed more labelling in these structures as well as in the cSNc. At 45 days after surgery comparable labelling was seen in both lesion groups. The total area of anterograde HRP-labelling in the iSNr significantly increased over time, with no differences between treatment groups. In Experiment 2, rats given the same hemitransections as in Experiment 1, were treated with daily injections of saline or GM1 for 14 days, and then received unilateral injections of 6-hydroxydopamine into the iSNc and iVTA. Nine days later, brain tissue was stained for examination of anterograde degeneration. Significantly more degenerating axons and terminals were found in the caudate nucleus of GM1-terminals were found in the caudate nucleus of GM1-treated rats than in saline-treated controls. We propose that the early reduction of behavioral deficits may be related to a ganglioside-induced reduction of secondary degeneration or edema. The effect of gangliosides on later behavioral recovery is to accelerate neuronal reorganization. This reorganization probably involves terminal proliferation of ascending, intact striatal afferents spared by the hemitransection.


Subject(s)
Corpus Striatum/injuries , Substantia Nigra/injuries , Animals , Apomorphine/pharmacology , Dextroamphetamine/pharmacology , G(M3) Ganglioside/therapeutic use , Hydroxydopamines/pharmacology , Male , Motor Activity/drug effects , Neural Pathways/injuries , Neuronal Plasticity/drug effects , Oxidopamine , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...