Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Int J Sports Med ; 37(4): 329-33, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26701829

ABSTRACT

An investigation of whether body water changes during the Giro d'Italia affected average maximal mean power (MMP) of different time durations and to establish whether phase-angle and body cell mass (BCM) are related to MMP in elite cyclists. Approximately 2 h after each stage of the race, a bioelectrical impedance analysis was performed on 8 cyclists and analysed according to bioelectrical impedance vector analyses. Additionally, MMP of different time durations were recorded during each stage. Body mass increased (p<0.001), vector-length shortened (p<0.001) and MMP15 (maximal mean power for 15 s; p=0.043) decreased in the course of the Giro d'Italia. The shortening of the vector was negatively related to MMP10 (r=- 0.749, p=0.032) and MMP15 (r=- 0.735, p=0.038) during stage 16 (heavy mountain-stage) and MMP60 (r=- 0.751, p=0.032), MMP300 (r=- 0.739, p=0.036) and MMP1800 (r=- 0.769, p=0.026) during stage 19 (time-trial). Additionally, the baseline phase-angle and BCM were associated to MMP15 best (r=0.781, p=0.022 and 0.756, p=0.030, respectively). In the course of the Giro d'Italia, MMP15 decreased, indicating progressive fatigue. The vector-length shortening and to some extent the body mass increase indicate that cyclists gained body water during the race. This gain was positively associated with performance during the last stages, possibly due to improved thermoregulation. Furthermore, phase-angle and BCM, shown to be linked to cellular function and to represent metabolic active tissue, reflect individual MMP of short duration in professional road cyclists.


Subject(s)
Athletes , Bicycling/physiology , Body Water/physiology , Adult , Body Weight , Electric Impedance , Humans , Young Adult
2.
Neurosurg Rev ; 37(1): 1-14, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24306170

ABSTRACT

Unruptured intracranial aneurysms represent a decisional challenge. Treatment risks have to be balanced against an unknown probability of rupture. A better understanding of the physiopathology is the basis for a better prediction of the natural history of an individual patient. Knowledge about the possible determining factors arises from a careful comparison between ruptured versus unruptured aneurysms and from the prospective observation and analysis of unbiased series with untreated, unruptured aneurysms. The key point is the correct identification of the determining variables for the fate of a specific aneurysm in a given individual. Thus, the increased knowledge of mechanisms of formation and eventual rupture of aneurysms should provide significant clues to the identification of rupture-prone aneurysms. Factors like structural vessel wall defects, local hemodynamic stress determined also by peculiar geometric configurations, and inflammation as trigger of a wall remodeling are crucial. In this sense the study of genetic modifiers of inflammatory responses together with the computational study of the vessel tree might contribute to identify aneurysms prone to rupture. The aim of this article is to underline the value of a unifying hypothesis that merges the role of geometry, with that of hemodynamics and of genetics as concerns vessel wall structure and inflammatory pathways.


Subject(s)
Aneurysm, Ruptured/etiology , Aneurysm/etiology , Intracranial Aneurysm/etiology , Aneurysm/genetics , Aneurysm/pathology , Aneurysm, Ruptured/genetics , Aneurysm, Ruptured/pathology , Environment , Hemodynamics , Humans , Intracranial Aneurysm/genetics , Intracranial Aneurysm/pathology , Risk Factors
3.
Clin Genet ; 83(1): 7-14, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22510019

ABSTRACT

Cerebral cavernous malformations (CCMs) are a diffuse cerebrovascular disease affecting approximately 0.5% of the population. A CCM is characterized by abnormally enlarged and leaky capillaries arranged in mulberry-like structures with no clear flow pattern. The lesion might predispose to seizures, focal neurological deficits or fatal intracerebral hemorrhage. However, a CCM can also remain neurologically silent. It might either occur sporadically or as an inherited disorder with incomplete penetrance and variable expressivity. Due to advances in imaging techniques, the incidence of CCM diagnoses are increasing, and the patient must be managed on a multidisciplinary basis: genetic counselling, treatment if needed, and follow-up. Advances have been made using radiological and pathological correlates of CCM lesions adding to the accumulated knowledge of this disease, although management of these patients is very variable among centers. This review is aimed at providing an update in genetic and molecular insights of this condition. Included are implications for genetic counselling, and possible approaches to prevention and treatment that derive from the understanding of pathogenetic mechanisms.


Subject(s)
Brain , Central Nervous System/pathology , Hemangioma, Cavernous, Central Nervous System , Microtubule-Associated Proteins , Proto-Oncogene Proteins , Brain/metabolism , Brain/pathology , Central Nervous System/metabolism , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/pathology , Genetic Counseling , Hemangioma, Cavernous, Central Nervous System/diagnosis , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/physiopathology , Hemangioma, Cavernous, Central Nervous System/therapy , Humans , KRIT1 Protein , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Molecular Targeted Therapy , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Seizures/genetics , Seizures/pathology
4.
Neuroscience ; 172: 12-9, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21044661

ABSTRACT

The positive effect of adult undifferentiated mesenchymal stem cells (MSCs) on neuronal survival has already been reported, although the mechanisms by which MSCs exert their effect are still a matter of debate. Here we have demonstrated that MSCs are able to prolong the survival of dorsal root ganglion (DRG) neurons mainly by inhibiting some proteolytic enzymes, and in particular the pathway of metalloproteinases (MMPs), a family of proteins that are involved in many neuronal processes, including survival. The inhibition of MMPs was both direct, by acting on MT-MMP1, and indirect, by acting on those proteins that regulate MMPs' activation, such as Timp-1 and Sparc. The importance of the MMPs' down-regulation for neuronal survival was also demonstrated by using N-isobutyl-N-(4-methoxyphenylsulfonyl)-glycyl hydroxamic acid (NNGH), a wide range inhibitor of metalloproteinases, which was able to increase the survival of DRG neurons in a significant manner. The down-regulation of MMPs, obtained both by MSC contact and by chemical inhibition, led to the inactivation of caspase 3, the executor of apoptotic death in DRG neurons cultured alone, while caspase 7 was found to be irrelevant for the apoptotic process. The capacity of MSCs to prevent apoptosis mainly by inactivating the metalloproteinase pathway is an important finding that sheds light on MSCs' mechanism of action, making undifferentiated MSCs a promising tool for the treatment of many different neurodegenerative pathologies.


Subject(s)
Ganglia, Spinal/enzymology , Mesenchymal Stem Cells/enzymology , Metalloproteases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Sensory Receptor Cells/enzymology , Signal Transduction/physiology , Animals , Apoptosis Regulatory Proteins/physiology , Cell Communication/drug effects , Cell Communication/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Hydroxamic Acids/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Metalloproteases/physiology , Neural Pathways/drug effects , Neural Pathways/enzymology , Neural Pathways/physiology , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Sulfonamides/pharmacology
6.
J Neuroimmunol ; 220(1-2): 104-7, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20149932

ABSTRACT

OBJECTIVE AND SUBJECTS: To examine in vivo levels of BAFF (B-cell activating factor of the tumor necrosis factor family) and APRIL (a proliferation-inducing ligand) in both the cerebrospinal fluid (CSF) and serum of 30 naïve MS patients and 79 subjects affected by acute or chronic inflammatory or non-inflammatory neurological diseases. DESIGN: Case-control study. RESULTS: No difference among groups was evidenced in serum BAFF or APRIL levels. By contrast, CSF levels of BAFF in MS (mean 144.3 pg/ml+/-141.2), although not significantly different from those observed in NIND (164.2 pg/ml+/-92.0), acute peripheral OIND (243.1 pg/ml+/-139.0) or chronic OIND (240.2 pg/ml+/-122.5), were significantly higher in acute central OIND patients (1274.0 pg/ml+/-803.8; p<0.001 vs. all groups). Similarly, CSF APRIL levels in MS (1541.0 pg/ml+/-1071.0), NIND (2629.0 pg/ml+/-1669.0), acute peripheral OIND (2834.0 pg/ml+/-1118.) or chronic OIND (2764.0 pg/ml+/-659.7) were not significantly different, while they were significantly higher in acute central OIND (6218.0 pg/ml+/-3790.0; p<0.001 vs. MS and NIND; and p<0.05 vs. acute peripheral OIND). CONCLUSIONS: Our results strongly suggest that further investigation is warranted to elucidate the role of BAFF and APRIL in MS and that serum levels of BAFF and APRIL do not reflect CSF levels.


Subject(s)
B-Cell Activating Factor/cerebrospinal fluid , Central Nervous System/immunology , Central Nervous System/metabolism , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/cerebrospinal fluid , Adult , Aged , B-Cell Activating Factor/analysis , B-Cell Activating Factor/blood , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers/analysis , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Case-Control Studies , Central Nervous System/physiopathology , Female , Humans , Immunity, Humoral/immunology , Lymphocyte Activation/immunology , Male , Middle Aged , Multiple Sclerosis/physiopathology , Predictive Value of Tests , Tumor Necrosis Factor Ligand Superfamily Member 13/analysis , Tumor Necrosis Factor Ligand Superfamily Member 13/blood , Up-Regulation/immunology
7.
Clin Neuropathol ; 28(4): 263-74, 2009.
Article in English | MEDLINE | ID: mdl-19642505

ABSTRACT

OBJECTIVE: Type A Niemann-Pick is a severe neurological disease, caused by a mutation of the gene of acid sphingomyelinase (ASM) and reduced enzyme activity. Some studies reported neuropathological changes occurring in the central nervous system of ASM deficient transgenic (ASMKO) mice, while a detailed study on the peripheral nervous system (PNS) at different ages is currently lacking. The aim of our study was to examine the pathological changes occurring in the PNS and in the spinal cord in an AMSKO model of Niemann-Pick disease (NPD) Type A. MATERIAL AND METHOD: Dorsal root ganglia (DRG), peripheral nerves and spinal cord specimens were obtained from ASMKO mice and age-matched wild type animals (age range = 1-7 months). They were observed at the light and electron microscope. Behavioral testing was performed to assess motor coordination and reactivity. Fluoro-Jade B was used as a high affinity fluorescent marker for degenerating neurons. RESULTS: Typical NPD cytoplasmic inclusions were observed in DRG neurons and satellite cells, in peripheral nerve Schwann cells, in spinal cord neurons and in endothelial cells. All these inclusions were present from the age of 1 month and increased with aging. By Fluoro-Jade B staining we demonstrated the occurrence of neuronal degeneration starting from 5 months of age. CONCLUSION: Despite the fact that a definite diagnosis of NPD Type A depends on enzymatic assays and/or molecular analysis, morphological investigation remains an important diagnostic procedure. Well-defined and complete neuropathological information about the ASMKO mouse model, inclusive of PNS examination, may be crucial in the pre-clinical evaluation of new therapies.


Subject(s)
Niemann-Pick Disease, Type A/pathology , Peripheral Nervous System/pathology , Spinal Cord/pathology , Animals , Disease Models, Animal , Fluoresceins , Mice , Mice, Transgenic , Organic Chemicals , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics
8.
Int J Immunopathol Pharmacol ; 21(1): 97-105, 2008.
Article in English | MEDLINE | ID: mdl-18336735

ABSTRACT

Human CD38 antigen is a 42-45 kDa type II transmembrane glycoprotein with a short N-terminal cytoplasmic domain and a long C-terminal extracellular region. It is widely expressed in different cell types including thymocytes, activated T cells, and terminally differentiated B cells (plasma cells) and it is involved in cellular proliferation and adhesion. CD38 acts as an ectocyclase that converts NAD+ to the Ca2+ -releasing second messenger cyclic ADP-ribose (cADPR). It has been also demonstrated that increased extracellular levels of NAD+ and cADPR are involved in inflammatory diseases and in cellular damage, such as ischemia. In the present study, we have characterized the expression of CD38 in human neuroblastoma SH-SY5Y cell line. All-trans-retinoic acid (ATRA) treatment was used to induce cell differentiation. Our results indicate that: a) even if SH-SY5Y cells have a negative phenotype express CD38 at nuclear level, ATRA treatment does not influence this pattern; b) CD38 localizing to the nucleus may co-localize with p80-coilin positive nuclear-coiled bodies; c) purified nuclei, by Western blot determinations using anti-CD38 antibodies, display a band with a molecular mass of approximately 42 kDa; d) SH-SY5Y cells show nuclear ADP-ribosyl cyclase due to CD38 activity; e) the basal level of CD38 mRNA shows a time-dependent increase after treatment with ATRA. These results suggest that the presence of constitutive fully functional CD38 in the SH-SY5Y nucleus has some important implications for intracellular generation of cADP-ribose and subsequent nucleoplasmic calcium release.


Subject(s)
ADP-ribosyl Cyclase 1/analysis , Membrane Glycoproteins/analysis , Neuroblastoma/chemistry , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/physiology , Cell Line, Tumor , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , RNA, Messenger/analysis , Tretinoin/pharmacology
9.
Curr Med Chem ; 15(6): 538-48, 2008.
Article in English | MEDLINE | ID: mdl-18336268

ABSTRACT

Neurodegenerative diseases do affect glial or neuronal cells in both the peripheral and central nervous systems. Although they are characterized by different features and a different onset, all the neurodegenerative diseases share the final steps that lead to cell death by apoptosis. Apoptosis occurs also during developmental neurogenesis. Neuron survival and differentiation depend on specific neurotrophic factors released by their targets. During degenerative diseases the loss of neuronal or glial cells is responsible for the disease's progression. Current therapies are focused on counteracting the degenerative events by acting on the molecular mechanisms involved in cellular death, or by the exogenous administration of pro-survival factors. The presence in many areas of both the peripheral and central nervous systems of niches of neural progenitors which can differentiate, under specific conditions, into neurons or glial cells opens up new therapeutic perspectives. The Mitogen-Activated Protein Kinase (MAPK) family, that includes ERK1/2, JNK/SAPK, p38 and ERK5, is involved in the survival, proliferation and differentiation of nervous cells. Some of the MAPKs promote the differentiation towards the neuron lineage, others towards the glial one. The MAPKs are also involved in apoptosis and may, therefore, play a role in neurodegeneration. This dual role of MAPKs may make it possible to design alternative and/or synergistic approaches to the treatment of degenerative diseases, either by using specific inhibitors of the MAPKs involved in apoptosis, or by increasing the activation of the MAPKs involved in neuronal survival and differentiation. The increased activation of pro-differentiative MAPKs can lead to the replacement of damaged neurons by undifferentiated progenitors and the slowing down of the disease's progression.


Subject(s)
Apoptosis/physiology , Cell Differentiation/physiology , Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
10.
Cytotherapy ; 9(5): 427-38, 2007.
Article in English | MEDLINE | ID: mdl-17786604

ABSTRACT

BACKGROUND: Periodontal disease is a degenerative illness that leads to resorption of the alveolar bone. Mesenchymal stromal cells (MSC) represent a novel tool for the production of biologic constructs for the treatment of degenerative bone diseases. The preparation of MSC differentiated into osteogenic lineage for clinical use requires the fulfillment of strict good manufacturing practice (GMP) procedures. METHODS: MSC were isolated from BM samples and then cultured under GMP conditions. MSC were characterized phenotypically and for their differentiative potential. Cells were seeded onto collagen scaffolds (Gingistat) and induced to differentiate into osteogenic lineages using clinical grade drugs compared with standard osteogenic supplements. Alizarin Red S stain was used to test the deposition of the mineral matrix. Standard microbiologic analysis was performed to verify the product sterility. RESULTS: The resulting MSC were negative for CD33, CD34 and HLA-DR but showed high expression of CD90, CD105 and HLA-ABC (average expressions of 94.3%, 75.8% and 94.2%, respectively). Chondrogenic, osteogenic and adipogenic differentiation potential was demonstrated. The MSC retained their ability to differentiate into osteogenic lineage when seeded onto collagen scaffolds after exposure to a clinical grade medium. Cell numbers and cell viability were adequate for clinical use, and microbiologic assays demonstrated the absence of any contamination. DISCUSSION: In the specific context of a degenerative bone disease with limited involvement of skeletal tissue, the combined use of MSC, exposed to an osteogenic clinical grade medium, and biomimetic biodegradable scaffolds offers the possibility of producing adequate numbers of biologic tissue-engineered cell-based constructs for use in clinical trials.


Subject(s)
Biocompatible Materials/pharmacology , Bone Regeneration/physiology , Bone Resorption/therapy , Mesenchymal Stem Cell Transplantation/methods , Periodontal Diseases/therapy , Stromal Cells/physiology , Absorbable Implants , Bone Density Conservation Agents/pharmacology , Bone Matrix/drug effects , Bone Matrix/metabolism , Bone Regeneration/drug effects , Bone Resorption/etiology , Bone Resorption/physiopathology , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Lineage/physiology , Cells, Cultured , Collagen/pharmacology , Guided Tissue Regeneration/methods , Humans , Jaw/pathology , Jaw/physiopathology , Osteoblasts/cytology , Osteoblasts/physiology , Periodontal Diseases/physiopathology , Stromal Cells/drug effects
13.
Arch Oral Biol ; 52(1): 64-73, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17049335

ABSTRACT

OBJECTIVE: Management of periodontal defects has always been a challenge in clinical periodontics. Recently mesenchymal stem cells (MSC) have been proposed for tissue regeneration in periodontal disease and repair of large bone defects. Bone regeneration has to be supported by a scaffold which has to be biocompatible, biodegradable, and able to support cell growth and differentiation. The aim of this study was to evaluate osteogenic differentiation of MSC seeded on a collagen scaffold. DESIGN: MSC were obtained from adult rat bone marrow, expanded and cultured in plastic dishes or seeded in a collagen scaffold (Gingistat). MSC were induced towards osteogenic differentiation using osteogenic supplements. Cell differentiation and calcium deposits were evaluated by immunoblotting, immunohistochemistry, histochemical techniques, enzymatic activity assay, and SEM-EDX analysis. Biomaterial in vitro degradation was evaluated by measuring mass reduction after incubation in culture medium. RESULTS: Rat MSC osteogenic differentiation was demonstrated by osteopontin and osteocalcin expression and an increase in alkaline phosphatase activity. MSC were distributed homogeneously in the collagen scaffold. Nodular aggregates and alizarin red stained calcium deposits were observed in MSC induced towards osteogenic differentiation cultured in dishes or seeded in the collagen scaffold. SEM-EDX analysis demonstrated that calcium co-localized with phosphorous. The biomaterial in vitro degraded in 4-5 weeks. CONCLUSIONS: MSC from bone marrow differentiate towards osteogenic lineage, representing a suitable cell source for bone formation in periodontal regeneration. Gingistat collagen scaffold supports MSC distribution and differentiation, but its short degradation time may be a limitation for a future application in bone tissue regeneration.


Subject(s)
Collagen/physiology , Mesenchymal Stem Cells/physiology , Osteogenesis/physiology , Alkaline Phosphatase/analysis , Animals , Anthraquinones/analysis , Bone Marrow Cells/physiology , Calcium/analysis , Cell Differentiation/physiology , Cells, Cultured , Coloring Agents/analysis , Female , Immunohistochemistry/methods , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/ultrastructure , Microscopy, Electron, Scanning , Osteocalcin/analysis , Osteopontin/analysis , Phosphorus/analysis , Rats , Rats, Sprague-Dawley
14.
Neurology ; 64(7): 1238-44, 2005 Apr 12.
Article in English | MEDLINE | ID: mdl-15824354

ABSTRACT

OBJECTIVE: To determine the influence of the APOE genotype on functional and cognitive outcome and on the incidence and prognosis of clinical vasospasm (delayed ischemic neurologic deficit [DIND]) in noncomatose patients with aneurysmal subarachnoid hemorrhage (SAH). METHODS: The authors reviewed the data of patients admitted for SAH to the Neurosurgical Departments of the San Gerardo Hospital of Monza (January 1996 to December 2001) and the Ospedali Riuniti of Bergamo (January 2002 to September 2003). The authors considered only noncomatose patients and evaluated outcome by means of the Rankin Disability Index and the Mini-Mental State Examination at least 6 months after the SAH. STATISTICAL ANALYSIS: Uni- and multivariate logistic regression. RESULTS: The authors included 101 patients. They found the epsilon4 allele in 26 patients (25.7%). The presence of the epsilon4 allele negatively affected the overall outcome (functional morbidity or cognitive morbidity, or both) (p = 0.0087) and, particularly, cognitive morbidity (p = 0.0028). Those with an epsilon4 allele had delayed ischemic neurologic deficit DINDs more frequently (p = 0.024) and, in the presence of DIND, they were more likely to show permanent neurologic deficits (p = 0.0051). CONCLUSIONS: ApoE4 negatively affects cognitive morbidity and delayed ischemic neurologic deficit recovery. The presence of an epsilon4 allele increases the risk of delayed ischemic neurologic deficit.


Subject(s)
Apolipoproteins E/genetics , Cognition Disorders/genetics , Genetic Predisposition to Disease/genetics , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/genetics , Vasospasm, Intracranial/genetics , Adult , Apolipoprotein E4 , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , DNA Mutational Analysis , Disease Progression , Female , Gene Frequency , Genotype , Humans , Male , Middle Aged , Neuropsychological Tests , Prognosis , Retrospective Studies , Subarachnoid Hemorrhage/physiopathology , Vasospasm, Intracranial/metabolism , Vasospasm, Intracranial/physiopathology
15.
Neurochem Int ; 46(3): 205-11, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15670636

ABSTRACT

In previous studies we demonstrated that resveratrol acts in an antiapoptotic manner on the paclitaxel-treated human neuroblastoma (HN) SH-SY5Y cell line inhibiting the apoptotic pathways induced by the antineoplastic drug. In the present study we evaluated the antiapoptotic effect of resveratrol, studying its activity on cell cycle progression. We determined the mitotic index of cultures exposed to resveratrol and paclitaxel alone or in combination, the cell cycle distribution by flow cytometric analysis (FACS), and the modulation of some relevant cell cycle regulatory proteins. Resveratrol is able to induce S-phase cell arrest and this interference with the cell cycle is associated with an increase of cyclin E and cyclin A, a downregulation of cyclin D1, and no alteration in cyclin B1 and cdk 1 activation. The resveratrol-induced S-phase block prevents SH-SY5Y from entering into mitosis, the phase of the cell cycle in which paclitaxel exerts its activity, explaining the antiapoptotic effect of resveratrol.


Subject(s)
Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Cycle/drug effects , Neuroblastoma/pathology , Paclitaxel/antagonists & inhibitors , Paclitaxel/pharmacology , Stilbenes/pharmacology , Brain Neoplasms/metabolism , Cell Division/drug effects , Cell Line, Tumor , Cyclin B/metabolism , Cyclin B1 , Cyclins/metabolism , Flow Cytometry , G2 Phase/drug effects , Humans , Immunoblotting , Maturation-Promoting Factor/metabolism , Mitosis/drug effects , Neuroblastoma/metabolism , Resveratrol
16.
J Biol Regul Homeost Agents ; 18(1): 9-17, 2004.
Article in English | MEDLINE | ID: mdl-15323355

ABSTRACT

Extra corporeal photochemotherapy (ECP) is an immunomodulating procedure used in several nonneurological diseases which, similarly to multiple sclerosis, are likely to be due to T-cell-mediated autoimmunity and it is probable that ECP can modulate the normal activity of peripheral blood mononuclear cells (PBMC). Using the Lewis rat experimental allergic encephalomyelitis (EAE) model of human multiple sclerosis (MS) we examined the effect of extracorporeal UV-A irradiation on psoralen-activated PBMC. In our experiment the comparison between the two groups of animals (ECP or sham-treatment) evidenced that the ECP treatment reduced the severity of EAE on clinical grounds and this result was confirmed by the pathological examination. The changes in the titers of anti-myelin antigen antibodies typical of EAE were also modulated by the procedure. Ex vivo examination evidenced a significant reduction in tumor-necrosis factor-alpha (TNF-alpha) released by PBMC after lipopolysaccharides (LPS) stimulation in culture. We conclude that ECP modifies the normal activity of PBMC during the course of EAE and it is possible that one of the anti-inflammatory mechanisms of action of ECP is correlated to a down-regulation of T-helper 1 lymphocytes activity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Leukocytes, Mononuclear/immunology , Animals , Corticosterone/metabolism , Cytokines/metabolism , Down-Regulation , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Humans , Light , Lipopolysaccharides/metabolism , Multiple Sclerosis/immunology , Myelin Basic Protein/metabolism , Photochemotherapy , Rats , Rats, Inbred Lew , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Ultraviolet Rays
17.
J Neuroimmunol ; 151(1-2): 55-65, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15145604

ABSTRACT

Pixantrone is less cardiotoxic and is similarly effective to mitoxantrone (MTX) as an antineoplastic drug. In our study, pixantrone reduced the severity of acute and decreased the relapse rate of chronic relapsing experimental allergic encephalomyelitis (EAE) in rats. A marked and long-lasting decrease in CD3+, CD4+, CD8+ and CD45RA+ blood cells and reduced anti-MBP titers were observed with both pixantrone and MTX. In vitro mitogen- and antigen-induced T-cell proliferation tests of human and rodents cells evidenced that pixantrone was effective at concentrations which can be effectively obtained after i.v. administration in humans. Cardiotoxicity was present only in MTX-treated rats. The effectiveness and the favorable safety profile makes pixantrone a most promising immunosuppressant agent for clinical use in multiple sclerosis (MS).


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunosuppressive Agents/therapeutic use , Isoquinolines/therapeutic use , T-Lymphocytes/drug effects , Acute Disease , Animals , Cell Division/drug effects , Cells, Cultured , Chronic Disease , Female , Humans , Immunosuppressive Agents/adverse effects , Isoquinolines/adverse effects , Lymphocyte Count , Mitoxantrone/adverse effects , Mitoxantrone/therapeutic use , Rats , T-Lymphocytes/immunology
18.
Neurochem Int ; 42(5): 419-29, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12510025

ABSTRACT

trans-Resveratrol (3,4',5-trihydroxystilbene) is able to significantly reduce paclitaxel-induced apoptosis in the human neuroblastoma (HN) SH-SY5Y cell line, acting on several cellular signaling pathways that are involved in paclitaxel-induced apoptosis. trans-Resveratrol reverses phosphorylation of Bcl-2 induced by paclitaxel and concomitantly blocks Raf-1 phosphorylation, also observed after paclitaxel exposure, thus suggesting that Bcl-2 inactivation may be dependent on the activation of the Raf/Ras cascade. trans-Resveratrol also reverses the sustained phosphorylation of JNK/SAPK, which specifically occurs after paclitaxel exposure.Overall, our observations demonstrate that (a) the toxic action of paclitaxel on neuronal-like cells is not only related to the effect of the drug on tubulin, but also to its capacity to activate several intracellular pathways leading to inactivation of Bcl-2, thus causing cells to die by apoptosis, (b) trans-resveratrol significantly reduces paclitaxel-induced apoptosis by modulating the cellular signaling pathways which commit the cell to apoptosis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Neuroblastoma/drug therapy , Paclitaxel/pharmacology , Signal Transduction/drug effects , Stilbenes/pharmacology , Brain Neoplasms/pathology , Caspase 7 , Caspases/biosynthesis , Caspases/genetics , Cell Death/drug effects , Cell Line , DNA/biosynthesis , DNA/genetics , Humans , Immunoblotting , Mitogen-Activated Protein Kinases/biosynthesis , Mitogen-Activated Protein Kinases/genetics , Neuroblastoma/pathology , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-raf/biosynthesis , Proto-Oncogene Proteins c-raf/genetics , Resveratrol , Tumor Cells, Cultured
19.
Neurosci Lett ; 322(2): 103-6, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11958854

ABSTRACT

The pathogenesis of the neurotoxicity of most antineoplastic drugs is unknown. Recent reports suggest that changes in the circulating levels of nerve growth factor (NGF) might be related to the dorsal root ganglia sensory neuron damage induced by cisplatin (CDDP), the first member of a family of widely used and very effective platinum-derived anticancer agents. Using a well-characterized model of CDDP neurotoxicity, we demonstrated that the NGF circulating level decreased during chronic CDDP administration in close accordance with the clinical course and returned to normal levels after recovery from the neurotoxic damage. Moreover, these changes were restricted to NGF and did not involve other trophic factors of the same neurotrophin family. Our findings are in agreement with previous in vitro and in vivo results and further suggest that NGF plays a specific role in the course of CDDP-induced primary sensory neuron damage.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Nerve Growth Factor/antagonists & inhibitors , Nerve Growth Factor/blood , Animals , Antineoplastic Agents/administration & dosage , Female , Injections, Intraperitoneal , Peripheral Nervous System/drug effects , Rats , Rats, Wistar
20.
Anticancer Res ; 22(6C): 4199-204, 2002.
Article in English | MEDLINE | ID: mdl-12553056

ABSTRACT

BACKGROUND: Oxaliplatin neurotoxicity represents a clinically-relevant problem and its etio-pathogenesis is still unknown. We explored the possible role of some neuronal growth factors ("neurotrophins") during the course of oxaliplatin sensory neuronopathy. MATERIALS AND METHODS: In our rat model two different doses of oxaliplatin were used (2 and 3 mg/kg i.v. twice weekly for 9 times). The neurotoxicity of the treatment was assessed with neurophysiological and pathological methods and serum neurotrophin levels were measured by ELISA. RESULTS: Both oxaliplatin-treated groups showed the neurophysiological and neuropathological changes which mimic the chronic effects of oxaliplatin administration in humans, e.g. reversible sensory impairment due to dorsal root ganglia neuron damage. These changes were associated with a significant and dose-dependent reduction only in the circulating level of nerve growth factor (NGF), which returned to normal values after neurophysiological and pathological recovery. CONCLUSION: This specific association between neurological impairment and NGF modulation indicates that NGF impairment has a role in the neurotoxicity of oxaliplatin.


Subject(s)
Antineoplastic Agents/toxicity , Nerve Growth Factors/blood , Organoplatinum Compounds/toxicity , Peripheral Nervous System Diseases/blood , Peripheral Nervous System Diseases/chemically induced , Animals , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Neural Conduction/drug effects , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Oxaliplatin , Peripheral Nervous System Diseases/pathology , Rats , Rats, Wistar , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Tail/innervation
SELECTION OF CITATIONS
SEARCH DETAIL
...