Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Int J Mol Sci ; 24(2)2023 Jan 14.
Article in English | MEDLINE | ID: mdl-36675203

ABSTRACT

Peripheral Neuropathies (PN) are common conditions whose treatment is still lacking in most cases. Animal models are crucial, but experimental procedures should be refined in some cases. We performed a detailed characterization of the ventral caudal nerve to contribute to a more effective assessment of axonal damage in future PN studies. PN was induced via weekly systemic injection of a neurotoxic drug (paclitaxel); we compared the control and PN-affected rats, performing serial neurophysiological evaluations of the caudal nerve for its entire length. On the same nerve portions, we performed light microscopy and ultrastructural pathological observations to assess the severity of damage and verify the integrity of the surrounding structures. Neurophysiological and morphological analyses confirmed that a severe axonopathy had ensued in the PN group, with a length-dependent modality, matching morphological observations. The site of neurophysiological recording (e.g., distance from the base of the tail) was critical for achieving useful data. A flexible experimental paradigm should be considered in animal studies investigating axonal PN, particularly if the expected severity is relevant; the mid-portion of the tail might be the most appropriate site: there damage might be remarkable but neither as extreme as at the tip of the tail nor as mild as at the base of the tail.


Subject(s)
Nerve Tissue , Neurotoxicity Syndromes , Peripheral Nervous System Diseases , Rats , Animals , Peripheral Nervous System Diseases/chemically induced , Nerve Tissue/pathology , Paclitaxel/adverse effects , Axons/pathology , Neurotoxicity Syndromes/pathology
2.
Int J Mol Sci ; 23(10)2022 May 21.
Article in English | MEDLINE | ID: mdl-35628600

ABSTRACT

Mesenchymal Stem Cells (MSCs) are adult multipotent cells able to increase sensory neuron survival: direct co-culture of MSCs with neurons is pivotal to observe a neuronal survival increase. Despite the identification of some mechanisms of action, little is known about how MSCs physically interact with neurons. The aim of this paper was to investigate and characterize the main mechanisms of interaction between MSCs and neurons. Morphological analysis showed the presence of gap junctions and tunneling nanotubes between MSCs and neurons only in direct co-cultures. Using a diffusible dye, we observed a flow from MSCs to neurons and further analysis demonstrated that MSCs donated mitochondria to neurons. Treatment of co-cultures with the gap junction blocker Carbenoxolone decreased neuronal survival, thus demonstrating the importance of gap junctions and, more in general, of cell communication for the MSC positive effect. We also investigated the role of extracellular vesicles; administration of direct co-cultures-derived vesicles was able to increase neuronal survival. In conclusion, our study demonstrates the presence and the importance of multiple routes of communication between MSCs and neurons. Such knowledge will allow a better understanding of the potential of MSCs and how to maximize their positive effect, with the final aim to provide the best protective treatment.


Subject(s)
Mesenchymal Stem Cells , Adult , Cell Communication , Cell Survival/physiology , Coculture Techniques , Humans , Sensory Receptor Cells
3.
Exp Cell Res ; 319(19): 2989-99, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23973667

ABSTRACT

Over the last few years the therapeutic approach to demyelinating diseases has radically changed, strategies having been developed aimed at partnering the classic symptomatic treatments with the most advanced regenerative medicine tools. At first, the transplantation of myelinogenic cells, Schwann cells or oligodendrocytes was suggested, but the considerable technical difficulties, (poor availability, difficulties in harvesting and culturing, and the problem of rejection in the event of non-autologous sources), shifted attention towards more versatile cellular types, such as Mesenchymal Stem Cells (MSCs). Recent studies have already demonstrate both in vitro and in vivo that glially-primed MSCs (through exposure to chemical cocktails) have myelogenic abilities. In spite of a large number of papers on glially-differentiated MSCs, little is known about the ability of undifferentiated MSCs to myelinate axons and processes. Here we have demonstrated that also undifferentiated MSCs have the ability to myelinate, since they induce the myelination of rat DRG neuron processes after direct co-culturing. In this process a pivotal role is performed by the p75 receptor.


Subject(s)
Cell Differentiation/physiology , Mesenchymal Stem Cells/metabolism , Myelin Sheath/physiology , Neurites/pathology , Neurons/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Axons/pathology , Cells, Cultured , Coculture Techniques , Nerve Tissue Proteins , Neurons/cytology , Oligodendroglia/cytology , Rats , Rats, Sprague-Dawley , Receptors, Growth Factor , Schwann Cells/cytology
4.
Toxics ; 11(2)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36850969

ABSTRACT

Chemotherapy-induced peripheral neurotoxicity is one of the most common dose-limiting toxicities of several widely used anticancer drugs such as platinum derivatives (cisplatin) and taxanes (paclitaxel). Several molecular mechanisms related to the onset of neurotoxicity have already been proposed, most of them having the sensory neurons of the dorsal root ganglia (DRG) and the peripheral nerve fibers as principal targets. In this study we explore chemotherapy-induced peripheral neurotoxicity beyond the neuronocentric view, investigating the changes induced by paclitaxel (PTX) and cisplatin (CDDP) on satellite glial cells (SGC) in the DRG and their crosstalk. Rats were chronically treated with PTX (10 mg/Kg, 1qwx4) or CDDP (2 mg/Kg 2qwx4) or respective vehicles. Morpho-functional analyses were performed to verify the features of drug-induced peripheral neurotoxicity. Qualitative and quantitative immunohistochemistry, 3D immunofluorescence, immunoblotting, and transmission electron microscopy analyses were also performed to detect alterations in SGCs and their interconnections. We demonstrated that PTX, but not CDDP, produces a strong activation of SGCs in the DRG, by altering their interconnections and their physical contact with sensory neurons. SGCs may act as principal actors in PTX-induced peripheral neurotoxicity, paving the way for the identification of new druggable targets for the treatment and prevention of chemotherapy-induced peripheral neurotoxicity.

5.
Med Phys ; 49(1): 568-578, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34778990

ABSTRACT

PURPOSE: We investigated the dose enhancement and internalization of gold nanoparticles (AuNPs) used as a radiosensitizer agent for rotational radiotherapy of breast cancer using a kilovoltage (kV) X-ray beam. METHODS: Human breast cancer cells MDA-MB-231 were incubated with or without 100Ā Āµg/mL (4.87Ā nM) or 200Ā Āµg/mL (9.74Ā nM) 15Ā nm AuNPs and irradiated with 100Ā kV, 190Ā kV, or 6 MV X-rays. To assess the toxicity of the AuNPs, we performed a Sulforhodamine B assay. Using atomic absorption spectroscopy, scanning electron microscopy, transmission electron microscopy, and time-lapse optical microscopy (rate of 2 frames per minute), we carried out a quantitative assessment of the amount of gold internalized by MDA-MB-231 cells and a characterization of the static and dynamical aspects of this internalization process. RESULTS: No effect of AuNPs alone was shown on cell viability. Time-lapse optical microscopy showed for the first time AuNPs cellular uptake and the dynamics of AuNPs internalization. Electron microscopy demonstrated AuNPs localization in endosomal vesicles, preferentially in the perinuclear region. After irradiation at doses up to 2Ā Gy, cell survival fraction curves showed increased mortality with AuNPs, with respect to irradiation without AuNPs. The highest effect of radioenhancement by AuNPs (at 9.74Ā nM AuNPs concentration) was observed at 190Ā kV showing a dose enhancement factor of 1.33Ā Ā±Ā 0.06 (1.34Ā Ā±Ā 0.02 at 100Ā kV), while at 6 MV it was 1.14Ā Ā±Ā 0.06. CONCLUSIONS: The observed radio-sensitization effect is promising for future radio-enhanced kV radiotherapy of breast cancer and quantitatively in the order of previous observations for 15Ā nm AuNPs. These results of a significant dose enhancement were obtained at 15Ā nm AuNPs concentration as low as several nanomolar units, at dose levels typical of a single dose fraction in a radiotherapy session. Dynamical behavior of the 3D spatial distribution of 15Ā nm AuNPs outside the nucleus of single breast cancer cell was observed, with possible implications for future models of AuNPs sensitization.


Subject(s)
Metal Nanoparticles , Radiation-Sensitizing Agents , Gold , Humans , Photons , Radiation-Sensitizing Agents/pharmacology , X-Rays
6.
Cancer Sci ; 101(2): 416-24, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19961489

ABSTRACT

Transitional cell carcinoma (TCC) is the most common type of bladder cancer. Emerging evidence has suggested that the capability of a tumor to grow and propagate is dependent on a small subset of cells, the cancer stem-like cells (CSCs) or tumor initiating cells. We report on the isolation and biological characterization of putative bladder CSC populations from primary TCCs. Isolated cells were induced to proliferate in stem cell culture conditions (serum-free medium containing mitogenic growth factors). The proliferating cells formed spheroids (urospheres) and their abilities for extensive proliferation and self-renewal were assayed. Their positivity for several stem cell markers (CD133, Oct-3/4, nestin, and cytokeratins) was also assessed by immunofluorescence tests and they could have the potential to differentiate in the presence of serum. In stem cell culture conditions they gradually showed loss of proliferation, adherence to the substrate, and morphological changes, which might reflect their progressive acquisition of differentiative capacity and loss of self-renewal ability. To evaluate if effective cell selection occurred after isolation, conventional cytogenetic studies on fresh chromosome spreads immediately after isolation and after culture were carried out. In addition, a molecular cytogenetic study by UroVysion assay was carried out on paraffin-embedded tissue sections and on fresh and after culture nuclei preparations. The data collected indicated important karyotype changes and a positive selection for hypo- or near-diploid cells, losing the complexity present in fresh tumors.


Subject(s)
Carcinoma, Transitional Cell/pathology , Neoplastic Stem Cells/pathology , Urinary Bladder Neoplasms/pathology , AC133 Antigen , Aged , Aged, 80 and over , Animals , Antigens, CD/analysis , Carcinoma, Transitional Cell/genetics , Cell Differentiation , Chromosome Aberrations , Female , Glycoproteins/analysis , Humans , Male , Mice , Middle Aged , Peptides/analysis , Urinary Bladder Neoplasms/genetics
7.
J Neurosci Methods ; 339: 108744, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32353471

ABSTRACT

BACKGROUND: Dense and unbiased cellular-resolution representations of extended volumetric central nervous system soft-tissue anatomy are difficult to obtain, even in experimental post-mortem settings. Interestingly, X-ray phase-contrast computed tomography (X-PCI-CT), an emerging soft-tissue-sensitive volumetric imaging technique, can provide multiscale organ- to cellular-level morphological visualizations of neuroanatomical structure. NEW METHOD: Here, we tested different nervous-tissue fixation procedures, conventionally used for transmission electron microscopy, to better establish X-PCI-CT-specific sample-preparation protocols. Extracted rat spinal medullas were alternatively fixed with a standard paraformaldehyde-only aldehyde-based protocol, or in combination with glutaraldehyde. Some specimens were additionally post-fixed with osmium tetroxide. Multiscale X-PCI-CT datasets were collected at several synchrotron radiation facilities, using state-of-the-art setups with effective image voxel sizes of 3.03 to 0.33 Āµm3, and compared to high-field magnetic resonance imaging, histology and vascular fluorescence microscopy data. RESULTS: Multiscale X-PCI-CT of aldehyde-fixed spinal cord specimens resulted in dense histology-like volumetric representations and quantifications of extended deep spinal micro-vascular networks and of intra-medullary cell populations. Osmium post-fixation increased intra-medullary contrast between white and gray-matter tissues, and enhanced delineation of intra-medullary cellular structure, e.g. axon fibers and motor neuron perikarya. COMPARISON WITH EXISTING METHODS: Volumetric X-PCI-CT provides complementary contrast and higher spatial resolution compared to 9.4Ć¢Ā€ĀÆT MRI. X-PCI-CT's advantage over planar histology is the volumetric nature of the cellular-level data obtained, using samples much larger than those fit for volumetric vascular fluorescence microscopy. CONCLUSIONS: Deliberately choosing (post-)fixation protocols tailored for optimal nervous-tissue structural preservation is of paramount importance in achieving effective and targeted neuroimaging via the X-PCI-CT technique.


Subject(s)
Osmium , Percutaneous Coronary Intervention , Aldehydes , Animals , Rats , Rodentia , Spinal Cord/diagnostic imaging , X-Ray Microtomography , X-Rays
8.
Nat Commun ; 11(1): 5938, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33230096

ABSTRACT

Recurrent somatic mutations in ETNK1 (Ethanolamine-Kinase-1) were identified in several myeloid malignancies and are responsible for a reduced enzymatic activity. Here, we demonstrate in primary leukemic cells and in cell lines that mutated ETNK1 causes a significant increase in mitochondrial activity, ROS production, and Histone H2AX phosphorylation, ultimately driving the increased accumulation of new mutations. We also show that phosphoethanolamine, the metabolic product of ETNK1, negatively controls mitochondrial activity through a direct competition with succinate at mitochondrial complex II. Hence, reduced intracellular phosphoethanolamine causes mitochondria hyperactivation, ROS production, and DNA damage. Treatment with phosphoethanolamine is able to counteract complex II hyperactivation and to restore a normal phenotype.


Subject(s)
Ethanolamines/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Cell Line , Cell Respiration/drug effects , Cell Respiration/genetics , DNA Breaks/drug effects , Electron Transport Complex II/drug effects , Electron Transport Complex II/metabolism , Ethanolamines/metabolism , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mitochondria/genetics , Mitochondria/pathology , Mutation , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Reactive Oxygen Species/metabolism , Succinic Acid/metabolism , Tigecycline/pharmacology
9.
J Anat ; 213(5): 539-46, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19014361

ABSTRACT

l-Glutamate is one of the major excitatory neurotransmitters in the mammalian central nervous system, but recently it has been shown to have a role also in the transduction of sensory input at the periphery, and in particular in the nociceptive pathway. An excess of glutamate is implicated in cases of peripheral neuropathies as well. Conventional therapeutic approaches for treating these diseases have focused on blocking glutamate receptors with small molecules or on reducing its synthesis of the receptors through the inhibition of glutamate carboxypeptidase II (GCPII), the enzyme that generates glutamate. In vivo studies have demonstrated that the pharmacological inhibition of GCPII can either prevent or treat the peripheral nerve changes in both BB/Wor and chemically induced diabetes in rats. In this study, we characterized the expression and distribution of glutamate transporters GLT1, GLAST, EAAC1 and of the enzyme GCPII in the peripheral nervous system of female Wistar rats. Immunoblotting results demonstrated that all glutamate transporters and GCPII are present in dorsal root ganglia (DRG) and the sciatic nerve. Immunofluorescence localization studies revealed that both DRG and sciatic nerves were immunopositive for all glutamate transporters and for GCPII. In DRG, satellite cells were positive for GLT1 and GCPII, whereas sensory neurons were positive for EAAC1. GLAST was localized in both neurons and satellite cells. In the sciatic nerve, GLT1 and GCPII were expressed in the cytoplasm of Schwann cells, whereas GLAST and EAAC1 stained the myelin layer. Our results give for the first time a complete characterization of the glutamate transporter system in the peripheral nervous system. Therefore, they are important both for understanding glutamatergic signalling in the PNS and for establishing new strategies to treat peripheral neuropathies.


Subject(s)
Excitatory Amino Acid Transporter 2/analysis , Peripheral Nervous System/metabolism , Animals , Biomarkers/analysis , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Excitatory Amino Acid Transporter 1/analysis , Excitatory Amino Acid Transporter 3/analysis , Female , Fluorescent Antibody Technique , Gene Expression , Glutamate Carboxypeptidase II/analysis , Microscopy, Confocal , Rats , Rats, Wistar , Sciatic Nerve/chemistry
10.
Anticancer Res ; 28(1A): 335-42, 2008.
Article in English | MEDLINE | ID: mdl-18383866

ABSTRACT

BACKGROUND: Antineoplastic drugs, such as cisplatin (CDDP), induce disabling peripheral neuropathies, representing a hindrance to effective cancer treatments. The exact pathogenesis of CDDP-induced neuropathy is not yet understood, and the dysregulation of gene expression has been proposed. Valproate (VPA) is an antiepileptic drug recently discovered to remodel gene expression, with hypothetically putative neuroprotective effects. MATERIALS AND METHODS: VPA was tested in both, in vitro and in vivo models of CDDP-neurotoxicity. RESULTS: VPA administered in combination with CDDP promoted dorsal root ganglia (DRG) neurons survival. Moreover, this treatment induced in Wistar rats an improvement of body weight, sensory nerve conduction velocity, and DRG morphometric analysis. In contrast, VPA was not able to rescue CDDP pre-treated rats. CONCLUSION: When used in combination with CDDP, VPA displays a protective action against neuropathy, in our models, suggesting possible future clinical applications.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/toxicity , Peripheral Nervous System Diseases/prevention & control , Valproic Acid/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/toxicity , Cisplatin/administration & dosage , Drug Synergism , Female , Ganglia, Spinal/drug effects , Neurites/drug effects , Peripheral Nervous System Diseases/chemically induced , Rats , Rats, Sprague-Dawley , Rats, Wistar , Valproic Acid/administration & dosage
11.
Anticancer Res ; 26(2A): 1065-70, 2006.
Article in English | MEDLINE | ID: mdl-16619507

ABSTRACT

Paclitaxel is an antineoplastic drug which acts by enhancing tubulin polymerization. The induction of peripheral neuropathy is the main dose-limiting side-effect of paclitaxel treatment. In this study, the neurotoxic effect of this drug in dorsal root ganglion (DRG) explants was analyzed by measuring the neurite length of DRG explants exposed to nerve growth factor (NGF). The neurotoxic effect of paclitaxel is dose- and time-dependent. Moreover, in DRG dissociated post-mitotic neurons, the molecular and morphological features of paclitaxel-induced cellular death were studied and the DRG neurons were observed to die by necrosis. On the contrary, the proliferating human neuroblastoma SH-SY5Y cells exposed to paclitaxel die by apoptosis, as reported for cortical neurons. The different response to the same stimulus of different neuronal populations underlines the importance of the biochemical and molecular phenotype of the neuronal population in determining cellular behavior and vulnerability to the same noxious stimulus.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Ganglia, Spinal/drug effects , Neurons/drug effects , Paclitaxel/toxicity , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Ganglia, Spinal/cytology , Neurites/drug effects , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Paclitaxel/pharmacology , Rats , Rats, Sprague-Dawley
12.
Neurobiol Aging ; 45: 136-148, 2016 09.
Article in English | MEDLINE | ID: mdl-27459934

ABSTRACT

This study is aimed at describing the changes occurring in the entire peripheral nervous system sensory pathway along a 2-year observation period in a cohort of C57BL/6 mice. The neurophysiological studies evidenced significant differences in the selected time points corresponding to childhood, young adulthood, adulthood, and aging (i.e., 1, 7, 15, and 25Ā months of age), with a parabolic course as function of time. The pathological assessment allowed to demonstrate signs of age-related changes since the age of 7Ā months, with a remarkable increase in both peripheral nerves and dorsal root ganglia at the subsequent time points. These changes were mainly in the myelin sheaths, as also confirmed by the Rotating-Polarization Coherent-Anti-stokes-Raman-scattering microscopy analysis. Evident changes were also present at the morphometric analysis performed on the peripheral nerves, dorsal root ganglia neurons, and skin biopsies. This extensive, multimodal characterization of the peripheral nervous system changes in aging provides the background for future mechanistic studies allowing the selection of the most appropriate time points and readouts according to the investigation aims.


Subject(s)
Aging/pathology , Aging/physiology , Neural Pathways/pathology , Neural Pathways/physiology , Peripheral Nervous System/pathology , Peripheral Nervous System/physiology , Animals , Female , Ganglia, Spinal/pathology , Ganglia, Spinal/physiology , Ganglia, Spinal/physiopathology , Mice, Inbred C57BL , Neural Conduction/physiology , Neural Pathways/physiopathology , Peripheral Nervous System/physiopathology , Skin/innervation
13.
J Neurosci ; 22(15): 6336-46, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12151511

ABSTRACT

Most CNS synapses investigated thus far contain a large number of vesicles docked at the active zone, possibly forming individual release sites. At the present time, it is unclear whether these vesicles can be discharged independently of one another. To investigate this problem, we recorded miniature excitatory currents by whole-cell and single-synapse recordings from CA3-CA1 hippocampal neurons and analyzed their stochastic properties. In addition, spontaneous release was investigated by ultrastructural analysis of quickly frozen synapses, revealing vesicle intermediates in docking and spontaneous fusion states. In these experiments, no signs of inhibitory interactions between quanta could be detected up to 1 msec from the previous discharge. This suggests that exocytosis at one site does not per se inhibit vesicular fusion at neighboring sites. At longer intervals, the output of quanta diverged from a random memoryless Poisson process because of the presence of a bursting component. The latter, which could not be accounted for by random coincidences, was independent of Ca2+ elevations in the cytosol, whether from Ca2+ flux through the plasma membrane or release from internal stores. Results of these experiments, together with the observation of spontaneous pairs of omega profiles at the active zone, suggest that multimodal release is produced by an enduring activation of an integrated cluster of release sites.


Subject(s)
Hippocampus/metabolism , Neural Inhibition/physiology , Synapses/metabolism , Synaptic Transmission/physiology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Cell Membrane/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Cytosol/metabolism , Endocytosis/physiology , Exocytosis/physiology , Freeze Fracturing , Hippocampus/drug effects , Hippocampus/ultrastructure , Membrane Fusion/physiology , Monte Carlo Method , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Patch-Clamp Techniques , Poisson Distribution , Presynaptic Terminals/metabolism , Rats , Reaction Time/physiology , Signal Processing, Computer-Assisted , Stochastic Processes , Synapses/drug effects , Synapses/ultrastructure , Synaptic Transmission/drug effects , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure
14.
Neurotoxicology ; 33(1): 1-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22079976

ABSTRACT

PURPOSE: Alterations in mRNA for myelin proteins are reported in animal models of chemotherapy-induced peripheral neuropathies (CIPN); however, ultrastructural changes in aldehyde-fixed and plastic-embedded myelin are not evident by electron microscopy. Therefore, we used X-ray diffraction (XRD) to investigate more subtle changes in myelin sheath structure from unfixed nerves. EXPERIMENTAL DESIGN: We used in vivo chronic animal models of CIPN in female Wistar rats, administering cisplatin (CDDP 2mg/kg, i.p. twice/week), paclitaxel (PT 10mg/kg, i.v. once/week) or bortezomib (0.20mg/kg, i.v. three times/week) over a total period of 4weeks. Animal weights were monitored, and tail nerve conduction velocity (NCV) was determined at the end of the treatments to assess the occurrence of peripheral neuropathy. Sciatic nerves were collected and the myelin structure was analyzed using electron microscopy (EM) and XRD. RESULTS: All the rats treated with the chemotherapy agents developed peripheral neuropathy, as indicated by a decrease in NCV values; however, light and electron microscopy indicated no severe pathological alterations of the myelin morphology. XRD also did not demonstrate significant differences between sciatic nerves in treated vs. control rats with respect to myelin period, relative amount of myelin, membrane structure, and regularity of membrane packing. CONCLUSIONS: These results indicate that experimental peripheral neuropathy caused by CDDP, PT, and bortezomib-which are among the most widely used chemotherapy agents-does not significantly affect the structure of internodal myelin in peripheral nerve.


Subject(s)
Antineoplastic Agents/toxicity , Myelin Proteins/metabolism , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/metabolism , Animals , Disease Models, Animal , Female , Microscopy, Electron, Transmission , Myelin Proteins/ultrastructure , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Neural Conduction/drug effects , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve/ultrastructure , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/physiopathology , Rats , Rats, Wistar , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sciatic Nerve/ultrastructure , X-Ray Diffraction
15.
PLoS One ; 5(11): e14035, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-21124963

ABSTRACT

Understanding the physiology of human neural stem cells (hNSCs) in the context of cell therapy for neurodegenerative disorders is of paramount importance, yet large-scale studies are hampered by the slow-expansion rate of these cells. To overcome this issue, we previously established immortal, non-transformed, telencephalic-diencephalic hNSCs (IhNSCs) from the fetal brain. Here, we investigated the fate of these IhNSC's immediate progeny (i.e. neural progenitors; IhNSC-Ps) upon unilateral implantation into the corpus callosum or the hippocampal fissure of adult rat brain, 3 days after global ischemic injury. One month after grafting, approximately one fifth of the IhNSC-Ps had survived and migrated through the corpus callosum, into the cortex or throughout the dentate gyrus of the hippocampus. By the fourth month, they had reached the ipsilateral subventricular zone, CA1-3 hippocampal layers and the controlateral hemisphere. Notably, these results could be accomplished using transient immunosuppression, i.e administering cyclosporine for 15 days following the ischemic event. Furthermore, a concomitant reduction of reactive microglia (Iba1+ cells) and of glial, GFAP+ cells was also observed in the ipsilateral hemisphere as compared to the controlateral one. IhNSC-Ps were not tumorigenic and, upon in vivo engraftment, underwent differentiation into GFAP+ astrocytes, and Ɵ-tubulinIII+ or MAP2+ neurons, which displayed GABAergic and GLUTAmatergic markers. Electron microscopy analysis pointed to the formation of mature synaptic contacts between host and donor-derived neurons, showing the full maturation of the IhNSC-P-derived neurons and their likely functional integration into the host tissue. Thus, IhNSC-Ps possess long-term survival and engraftment capacity upon transplantation into the globally injured ischemic brain, into which they can integrate and mature into neurons, even under mild, transient immunosuppressive conditions. Most notably, transplanted IhNSC-P can significantly dampen the inflammatory response in the lesioned host brain. This work further supports hNSCs as a reliable and safe source of cells for transplantation therapy in neurodegenerative disorders.


Subject(s)
Brain Ischemia/surgery , Graft Survival/immunology , Immunocompromised Host/immunology , Neural Stem Cells/transplantation , Animals , Brain Ischemia/pathology , Cell Movement/immunology , Cell Survival/immunology , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Corpus Callosum/metabolism , Corpus Callosum/pathology , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunohistochemistry , Male , Microglia/metabolism , Microglia/pathology , Microscopy, Electron , Neural Stem Cells/metabolism , Neural Stem Cells/ultrastructure , Rats , Rats, Sprague-Dawley , Stem Cell Transplantation/methods , Time Factors , Transplantation, Heterologous
17.
Eur J Cancer ; 45(4): 656-65, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19091544

ABSTRACT

We have characterised for the first time the general and neurological side effects experienced when using a series of chronic non-lethal cisplatin + paclitaxel schedules in Wistar rats, selected according to our previous experience and the animals' maximum tolerated dose. At the pathological level, the use of combination schedules was definitely more toxic at the kidney and sternal bone marrow level than the single-agent schedules. At the neurophysiological examination based on the assessment of the nerve conduction velocity measurement in the tail nerve, we identified only one combination schedule that was more neurotoxic than the similar schedules based on single-agent administration. This observation was confirmed by the neuropathological examination performed on the sciatic nerve, dorsal root ganglia, ventral and dorsal roots. Our study supports the hypothesis that the general and, to a lesser extent, neurological effects of a combination of cisplatin and paclitaxel are different from those of the administration of both drugs as single agents. We believe that these models may be useful for testing neuroprotective strategies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/toxicity , Neurotoxicity Syndromes/etiology , Peripheral Nervous System Diseases/chemically induced , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bone Marrow Diseases/chemically induced , Bone Marrow Diseases/pathology , Cisplatin/administration & dosage , Cisplatin/toxicity , Disease Models, Animal , Drug Administration Schedule , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Neurotoxicity Syndromes/pathology , Paclitaxel/administration & dosage , Paclitaxel/toxicity , Peripheral Nervous System Diseases/pathology , Rats , Rats, Wistar , Weight Loss/drug effects
18.
Neurotoxicology ; 30(2): 312-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19428505

ABSTRACT

An unsolved question is how platinum derivatives used for solid cancer therapy cause peripheral neuropathy in patients and apoptosis in "in vitro" models of chemotherapy-induced peripheral neuropathy. DRG neurons from E15 rat embryos were treated with toxic doses of oxaliplatin or cisplatin. Here, the role of MAPKs in neuronal apoptosis was studied. Both oxaliplatin and cisplatin induced a dose-dependent neuronal apoptosis, modulated by the proteins of Bcl-2 family. Regarding MAPKs, platinum derivatives activated p38 while they reduced the active form and the total amount of JNK/Sapk. Both oxaliplatin and cisplatin activated ERKs at early stages, although they behaved differently at later stages. By using specific inhibitors of the various MAPKs it was demonstrated that the platinum-induced neuronal apoptosis is mediated by early p38 and ERK1/2 activation, while JNK/Sapk has a neuroprotective role. These results suggest a role for the different MAPKs in peripheral neuropathies characterized by apoptosis of DRG neurons.


Subject(s)
Apoptosis/drug effects , Mitogen-Activated Protein Kinases/physiology , Neurons/drug effects , Neurons/enzymology , Platinum/toxicity , Animals , Apoptosis/physiology , Cells, Cultured , Female , Neurons/cytology , Pregnancy , Rats , Rats, Sprague-Dawley
19.
Exp Neurol ; 204(1): 317-25, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17214983

ABSTRACT

Bortezomib is a new proteasome inhibitor with a high antitumor activity, but also with a potentially severe peripheral neurotoxicity. To establish a preclinical model and to characterize the changes induced on the peripheral nerves, dorsal root ganglia (DRG) and spinal cord, bortezomib was administered to Wistar rats (0.08, 0.15, 0.20, 0.30 mg/kg/day twice [2q7d] or three times [3q7d] weekly for a total of 4 weeks). At baseline, on days 14, 21 and 28 after the beginning the treatment period and during a 4-week follow-up period sensory nerve conduction velocity (SNCV) was determined in the tail of each animal. Sciatic nerve, DRG and spinal cord specimens were processed for light and electron microscope observations and morphometry. At the maximum tolerated dose bortezomib induced a significant reduction in SNCV, with a complete recovery at the end of the follow-up period. Sciatic nerve examination and morphometric determinations demonstrated mild to moderate pathological changes, involving predominantly the Schwann cells and myelin, although axonal degeneration was also observed. Bortezomib-induced changes were also observed in DRG and they were represented by satellite cell intracytoplasmatic vacuolization due to mitochondrial and endoplasmic reticulum damage, closely resembling the changes observed in sciatic nerve Schwann cells. Only rarely did the cytoplasm of DRG neurons has a dark appearance and clear vacuoles occurring in the cytoplasm. Spinal cord was morphologically normal. This model is relevant to the neuropathy induced by bortezomib in the treatment of human malignancies and it could be useful in increasing our knowledge regarding the mechanisms underlying bortezomib neurotoxicity.


Subject(s)
Boronic Acids , Neurotoxins , Peripheral Nervous System Diseases/chemically induced , Pyrazines , Animals , Boronic Acids/toxicity , Bortezomib , Female , Ganglia, Spinal/pathology , Microscopy, Electron , Neural Conduction , Neurons, Afferent , Neurotoxins/toxicity , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/physiopathology , Pyrazines/toxicity , Rats , Rats, Wistar , Sciatic Nerve/pathology , Spinal Cord/pathology , Tail/innervation
20.
Proc Natl Acad Sci U S A ; 102(30): 10634-9, 2005 Jul 26.
Article in English | MEDLINE | ID: mdl-16027360

ABSTRACT

Junctional Adhesion Molecule-A (JAM-A) is a transmembrane adhesive protein expressed at endothelial junctions and in leukocytes. Here we report that JAM-A is required for the correct infiltration of polymorphonuclear leukocytes (PMN) into an inflamed peritoneum or in the heart upon ischemia-reperfusion injury. The defect was not observed in mice with an endothelium-restricted deficiency of the protein but was still detectable in mice transplanted with bone marrow from JAM-A(-/-) donors. Microscopic examination of mesenteric and heart microvasculature of JAM-A(-/-) mice showed high numbers of PMN adherent on the endothelium or entrapped between endothelial cells and the basement membrane. In vitro, in the absence of JAM-A, PMN adhered more efficiently to endothelial cells and basement membrane proteins, and their polarized movement was strongly reduced. This paper describes a nonredundant role of JAM-A in controlling PMN diapedesis through the vessel wall.


Subject(s)
Cell Adhesion Molecules/deficiency , Neutrophils/metabolism , Peritonitis/metabolism , Reperfusion Injury/metabolism , Animals , Bone Marrow Transplantation , Cell Adhesion/physiology , Cell Movement/physiology , Endothelium, Vascular/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Electron , Neutrophils/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL