Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Sci Adv ; 10(22): eadn2050, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38809982

ABSTRACT

Transporting and translating mRNAs in axons is crucial for neuronal viability. Local synthesis of nuclear-encoded mitochondrial proteins protects long-lived axonal mitochondria from damage; however, the regulatory factors involved are largely unknown. We show that CLUH, which binds mRNAs encoding mitochondrial proteins, prevents peripheral neuropathy and motor deficits in the mouse. CLUH is enriched in the growth cone of developing spinal motoneurons and is required for their growth. The lack of CLUH affects the abundance of target mRNAs and the corresponding mitochondrial proteins more prominently in axons, leading to ATP deficits in the growth cone. CLUH interacts with ribosomal subunits, translation initiation, and ribosome recycling components and preserves axonal translation. Overexpression of the ribosome recycling factor ABCE1 rescues the mRNA and translation defects, as well as the growth cone size, in CLUH-deficient motoneurons. Thus, we demonstrate a role for CLUH in mitochondrial quality control and translational regulation in axons, which is essential for their development and long-term integrity and function.


Subject(s)
Axons , Mitochondria , Motor Neurons , Peripheral Nervous System Diseases , Protein Biosynthesis , Animals , Motor Neurons/metabolism , Mitochondria/metabolism , Axons/metabolism , Mice , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/pathology , Growth Cones/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Mice, Knockout
2.
J Neuroimmune Pharmacol ; 19(1): 26, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38819756

ABSTRACT

BACKGROUND: The composition of gut microbiota plays a pivotal role in priming the immune system and thus impacts autoimmune diseases. Data on the effects of gut bacteria eradication via systemic antibiotics on immune neuropathies are currently lacking. This study therefore assessed the effects of antibiotics-induced gut microbiota alterations on the severity of experimental autoimmune neuritis (EAN), a rat model of Guillain-Barré Syndrome (GBS). Myelin P0 peptide 180-199 (P0 180-199)-induced EAN severity was compared between adult Lewis rats (12 weeks old) that received drinking water with or without antibiotics (colistin, metronidazole, vancomycin) and healthy rats, beginning antibiotics treatment immediately after immunization (day 0), and continuing treatment for 14 consecutive days. Neuropathy severity was assessed via a modified clinical score, and then related to gut microbiota alterations observed after fecal 16S rRNA gene sequencing at baseline and after EAN induction. Effectors of gut mucosal and endoneurial immunity were assessed via immunostaining. EAN rats showed increased gut mucosal permeability alongside increased mucosal CD8+ T cells compared to healthy controls. Antibiotics treatment alleviated clinical EAN severity and reduced endoneurial T cell infiltration, decreased gut mucosal CD8+ T cells and increased gut bacteria that may be associated with anti-inflammatory mechanisms, like Lactobacillus or Parasutterella. Our findings point out a relation between gut mucosal immunity and the pathogenesis of EAN, and indicate that antibiotics-induced intestinal immunomodulation might be a therapeutic approach to alleviate autoimmunity in immune neuropathies. Further studies are warranted to evaluate the clinical transferability of these findings to patients with GBS.


Subject(s)
Anti-Bacterial Agents , Gastrointestinal Microbiome , Immunomodulation , Neuritis, Autoimmune, Experimental , Rats, Inbred Lew , Animals , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/drug therapy , Rats , Gastrointestinal Microbiome/drug effects , Anti-Bacterial Agents/pharmacology , Immunomodulation/drug effects , Male
3.
Eur J Neurol ; 31(1): e16079, 2024 01.
Article in English | MEDLINE | ID: mdl-37789648

ABSTRACT

BACKGROUND AND PURPOSE: It is not known whether the route of administration affects the mechanisms of action of therapeutic immunoglobulin in chronic inflammatory demyelinating polyneuropathy (CIDP). The aim of this study, therefore, was to compare the immunomodulatory effects of intravenous (IVIg) and subcutaneous immunoglobulin (SCIg) in patients with CIDP and in IVIg-treated common variable immunodeficiency (CVID) patients. METHODS: Serum and peripheral blood mononuclear cell samples were obtained from 30 CIDP patients receiving IVIg, 10 CIDP patients receiving SCIg, and 15 patients with CVID receiving IVIg. Samples and clinical data were obtained prior to IVIg/SCIg and at 3 days, 7 days, and, in CIDP patients receiving IVIg, 21 days post-administration. Serum cytokines were assessed by Luminex-based multiplex assay and enzyme-linked immunosorbent assay. Immune cells were characterized by flow cytometry. RESULTS: Immune cell profiles of CIDP and CVID patients differed in frequencies of myeloid dendritic cells and cytotoxic natural killer cells. During treatment with IVIg or SCIg in CIDP patients, cellular immunomarkers were largely similar. CIDP patients receiving IVIg had higher macrophage inflammatory protein (MIP)-1α (p = 0.01), interleukin (IL)-4 (p = 0.04), and IL-33 (p = 0.04) levels than SCIg recipients. IVIg treatment more broadly modulated cytokines in CIDP than SCIg treatment. CONCLUSIONS: Our study demonstrates that the modulation of cellular immunomarkers in CIDP is independent of the application route of therapeutic immunoglobulin. Minor differences were observed between CIDP and CVID patients. In contrast, cytokines were differentially modulated by IVIg and SCIg in CIDP.


Subject(s)
Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Humans , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/drug therapy , Immunoglobulins, Intravenous/therapeutic use , Leukocytes, Mononuclear , Administration, Intravenous , Cytokines
4.
Neurosci Lett ; 814: 137456, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37648059

ABSTRACT

OBJECTIVE: We evaluated the potential neuro-regenerative effects of the mitochondrial uncoupler 2,4-Dinitrophenol in experimental autoimmune neuritis, an animal model for an acute autoimmune neuropathy. METHODS: Experimental autoimmune neuritis was induced in Lewis rats. Different concentrations of 2,4-Dinitrophenol (1 mg/kg, 0.1 mg/kg and 0.01 mg/kg) were applied during the recovery phase of the neuritis (at days 18, 22 and 26) and compared to the vehicle. Any effects were assessed through functional, electrophysiological, and morphological analysis via electron microscopy of all groups at day 30. Additional immune-histochemical analysis of inflammation markers and remyelination of the sciatic nerves were performed for the dosage of 1 mg/kg and control. RESULTS: No enhancement of functional or electrophysiological recovery was observed in all 2,4-Dinitrophenol-treated groups. Cellular inflammation markers of T cells (CD3+) were comparable to control, and an increase of macrophages (IbA1+) invasion in the sciatic nerves was observed. Treatment with 2,4-Dinitrophenol reduced axonal swelling in myelinated and unmyelinated fibers with an increased production of brain-derived neurotrophic factor. CONCLUSION: Our findings do not support the hypothesis that repurposing of the mitochondrial uncoupler 2,4-Dinitrophenol exerts functionally relevant neuro-regenerative effects in autoimmune neuritis.


Subject(s)
Neuritis, Autoimmune, Experimental , Neuritis , Rats , Animals , Rats, Inbred Lew , Neuritis, Autoimmune, Experimental/drug therapy , 2,4-Dinitrophenol/pharmacology , Dinitrophenols , Inflammation
5.
J Neuroinflammation ; 20(1): 139, 2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37296476

ABSTRACT

BACKGROUND: Autoimmune neuropathies can result in long-term disability and incomplete recovery, despite adequate first-line therapy. Kinesin-5 inhibition was shown to accelerate neurite outgrowth in different preclinical studies. Here, we evaluated the potential neuro-regenerative effects of the small molecule kinesin-5 inhibitor monastrol in a rodent model of acute autoimmune neuropathies, experimental autoimmune neuritis. METHODS: Experimental autoimmune neuritis was induced in Lewis rats with the neurogenic P2-peptide. At the beginning of the recovery phase at day 18, the animals were treated with 1 mg/kg monastrol or sham and observed until day 30 post-immunisation. Electrophysiological and histological analysis for markers of inflammation and remyelination of the sciatic nerve were performed. Neuromuscular junctions of the tibialis anterior muscles were analysed for reinnervation. We further treated human induced pluripotent stem cells-derived secondary motor neurons with monastrol in different concentrations and performed a neurite outgrowth assay. RESULTS: Treatment with monastrol enhanced functional and histological recovery in experimental autoimmune neuritis. Motor nerve conduction velocity at day 30 in the treated animals was comparable to pre-neuritis values. Monastrol-treated animals showed partially reinnervated or intact neuromuscular junctions. A significant and dose-dependent accelerated neurite outgrowth was observed after kinesin-5 inhibition as a possible mode of action. CONCLUSION: Pharmacological kinesin-5 inhibition improves the functional outcome in experimental autoimmune neuritis through accelerated motor neurite outgrowth and histological recovery. This approach could be of interest to improve the outcome of autoimmune neuropathy patients.


Subject(s)
Induced Pluripotent Stem Cells , Neuritis, Autoimmune, Experimental , Rats , Animals , Humans , Neuritis, Autoimmune, Experimental/drug therapy , Neuritis, Autoimmune, Experimental/pathology , Kinesins/therapeutic use , Rats, Inbred Lew , Induced Pluripotent Stem Cells/pathology
6.
Cells ; 12(9)2023 04 25.
Article in English | MEDLINE | ID: mdl-37174648

ABSTRACT

Peripheral neuropathy is a common side effect of cancer treatment with paclitaxel. The mechanisms by which paclitaxel is transported into neurons, which are essential for preventing neuropathy, are not well understood. We studied the uptake mechanisms of paclitaxel into neurons using inhibitors for endocytosis, autophagy, organic anion-transporting polypeptide (OATP) drug transporters, and derivatives of paclitaxel. RT-qPCR was used to investigate the expression levels of OATPs in different neuronal tissues and cell lines. OATP transporters were pharmacologically inhibited or modulated by overexpression and CRISPR/Cas9-knock-out to investigate paclitaxel transport in neurons. Through these experiments, we identified OATP1A1 and OATP1B2 as the primary neuronal transporters for paclitaxel. In vitro inhibition of OATP1A1 and OAT1B2 by glycyrrhizic acid attenuated neurotoxicity, while paclitaxel's antineoplastic effects were sustained in cancer cell lines. In vivo, glycyrrhizic acid prevented paclitaxel-induced toxicity and improved behavioral and electrophysiological measures. This study indicates that a set of OATPs are involved in paclitaxel transport into neurons. The inhibition of OATP1A1 and OATP1B2 holds a promising strategy to prevent paclitaxel-induced peripheral neuropathy.


Subject(s)
Organic Anion Transporters , Peripheral Nervous System Diseases , Humans , Paclitaxel/adverse effects , Glycyrrhizic Acid/pharmacology , Organic Anion Transporters/metabolism , Neurons/metabolism , Membrane Transport Proteins , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/prevention & control
7.
J Neuroimmune Pharmacol ; 18(1-2): 208-214, 2023 06.
Article in English | MEDLINE | ID: mdl-36929282

ABSTRACT

Availability of COVID-19 mRNA vaccine for patients with chronic inflammatory demyelinating polyneuropathy (CIDP) treated with intravenous immunoglobulin (IVIg) raises the question of whether COVID-19 mRNA vaccine influences disease activity or IVIg-mediated immunomodulation in CIDP. In this exploratory study, blood samples of CIDP patients on IVIg treatment were longitudinally analyzed before and after vaccination with a COVID-19 mRNA vaccine. A total of 44 samples of eleven patients were characterized at four timepoints by ELISA and flow cytometry in terms of immunomarkers for disease activity and IVIg-immunomodulation. Apart from a significantly lower expression of CD32b on naïve B cells after vaccination, no significant alteration of immunomarkers for CIDP or IVIg-mediated immunomodulation was observed. Our exploratory study suggests that COVID-19 mRNA vaccine does not have a relevant impact on immune activity in CIDP. In addition, immunomodulatory effects of IVIg in CIDP are not altered by COVID-19 mRNA vaccine. This study was registered in the German clinical trial register (DRKS00025759). Overview over the study design. Blood samples of CIDP patients on recurrent IVIg treatment and vaccination with a COVID-19 mRNA vaccine were obtained at four timepoints for cytokine ELISA and flow cytometry, to assess key cytokines and cellular immunomarkers for disease activity and IVIg-immunomodulation in CIDP.


Subject(s)
COVID-19 , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Humans , Immunoglobulins, Intravenous/therapeutic use , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/drug therapy , COVID-19 Vaccines , Vaccination , RNA, Messenger/therapeutic use
8.
Eur J Neurol ; 30(11): 3551-3556, 2023 11.
Article in English | MEDLINE | ID: mdl-36651357

ABSTRACT

BACKGROUND AND PURPOSE: The gut microbiome is involved in autoimmunity. Data on its composition in chronic inflammatory demyelinating polyneuropathy (CIDP), the most common chronic autoimmune disorder of peripheral nerves, are currently lacking. METHODS: In this monocentric exploratory pilot study, stool samples were prospectively collected from 16 CIDP patients (mean age 58 ± 10 years, 25% female) before and 1 week after administration of intravenous immunoglobulin (IVIg). Gut microbiota were analyzed via bacterial 16S rRNA gene sequencing and compared to 15 age-matched healthy subjects (mean age 59 ± 15 years, 66% female). RESULTS: The gut microbiota of CIDP patients showed an increased alpha-diversity (p = 0.005) and enrichment of Firmicutes, such as Blautia (p = 0.0004), Eubacterium hallii (p = 0.0004), or Ruminococcus torques (p = 0.03), and of Actinobacteriota (p = 0.03) compared to healthy subjects. IVIg administration did not alter the gut microbiome composition in CIDP in this short-term observation (p = 0.95). CONCLUSIONS: The gut microbiome in IVIg-treated CIDP shows distinct features, with increased bacterial diversity and enrichment of short-chain fatty acid producing Firmicutes. IVIg had no short-term impact on the gut microbiome in CIDP patients. As the main limitation of this exploratory pilot study was small cohort size, future studies also including therapy-naïve patients are warranted to verify our findings and to explore the impact of long-term IVIg treatment on the gut microbiome in CIDP.


Subject(s)
Gastrointestinal Microbiome , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Humans , Female , Middle Aged , Aged , Adult , Male , Immunoglobulins, Intravenous/therapeutic use , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/therapy , Pilot Projects , RNA, Ribosomal, 16S/genetics , Chronic Disease
9.
Front Pharmacol ; 13: 1038285, 2022.
Article in English | MEDLINE | ID: mdl-36408236

ABSTRACT

Glia are critical players in defining synaptic contacts and maintaining neuronal homeostasis. Both astrocytes as glia of the central nervous system (CNS), as well as satellite glial cells (SGC) as glia of the peripheral nervous system (PNS), intimately interact with microglia, especially under pathological conditions when glia regulate degenerative as well as regenerative processes. The chemotherapeutic agent paclitaxel evokes peripheral neuropathy and cognitive deficits; however, the mechanisms underlying these diverse clinical side effects are unclear. We aimed to elucidate the direct effects of paclitaxel on the function of astrocytes, microglia, and SGCs, and their glia-glia and neuronal-glia interactions. After intravenous application, paclitaxel was present in the dorsal root ganglia of the PNS and the CNS of rodents. In vitro, SGC enhanced the expression of pro-inflammatory factors and reduced the expression of neurotrophic factor NT-3 upon exposure to paclitaxel, resulting in predominantly neurotoxic effects. Likewise, paclitaxel induced a switch towards a pro-inflammatory phenotype in microglia, exerting neurotoxicity. In contrast, astrocytes expressed neuroprotective markers and increasingly expressed S100A10 after paclitaxel exposure. Astrocytes, and to a lesser extent SGCs, had regulatory effects on microglia independent of paclitaxel exposure. Data suggest that paclitaxel differentially modulates glia cells regarding their (neuro-) inflammatory and (neuro-) regenerative properties and also affects their interaction. By elucidating those processes, our data contribute to the understanding of the mechanistic pathways of paclitaxel-induced side effects in CNS and PNS.

10.
Eur J Neurol ; 29(11): 3380-3388, 2022 11.
Article in English | MEDLINE | ID: mdl-35842740

ABSTRACT

BACKGROUND AND PURPOSE: This study assessed the prevalence of anti-SARS-CoV-2 antibodies in therapeutic immunoglobulin and their impact on serological response to COVID-19 mRNA vaccine in patients with intravenous immunoglobulin (IVIg)-treated chronic immune neuropathies. METHODS: Forty-six samples of different brands or lots of IVIg or subcutaneous IgG were analyzed for anti-SARS-CoV-2 IgG using enzyme-linked immunosorbent assay and chemiluminescent microparticle immunoassay. Blood sera from 16 patients with immune neuropathies were prospectively analyzed for anti-SARS-CoV-2 IgA, IgG, and IgM before and 1 week after IVIg infusion subsequent to consecutive COVID-19 mRNA vaccine doses and after 12 weeks. These were compared to 42 healthy subjects. RESULTS: Twenty-four (52%) therapeutic immunoglobulin samples contained anti-SARS-CoV-2 IgG. All patients with immune neuropathies (mean age = 65 ± 16 years, 25% female) were positive for anti-SARS-CoV-2 IgG after COVID-19 vaccination. Anti-SARS-CoV-2 IgA titers significantly decreased 12-14 weeks after vaccination (p = 0.02), whereas IgG titers remained stable (p = 0.2). IVIg did not significantly reduce intraindividual anti-SARS-CoV-2 IgA/IgG serum titers in immune neuropathies (p = 0.69). IVIg-derived anti-SARS-CoV-2 IgG did not alter serum anti-SARS-CoV-2 IgG decrease after IVIg administration (p = 0.67). CONCLUSIONS: Our study indicates that IVIg does not impair the antibody response to COVID-19 mRNA vaccine in a short-term observation, when administered a minimum of 2 weeks after each vaccine dose. The infusion of current IVIg preparations that contain anti-SARS-CoV-2 IgG does not significantly alter serum anti-SARS-CoV-2 IgG titers.


Subject(s)
COVID-19 , Severe acute respiratory syndrome-related coronavirus , Aged , Aged, 80 and over , Antibodies, Viral , Antibody Formation , COVID-19 Vaccines , Female , Humans , Immunoglobulin A , Immunoglobulin G , Immunoglobulin M , Immunoglobulins, Intravenous/therapeutic use , Male , Middle Aged , SARS-CoV-2 , Vaccination , Vaccines, Synthetic , mRNA Vaccines
11.
Toxics ; 9(10)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34678925

ABSTRACT

Peripheral neuropathy is one of the most common side effects of chemotherapy, affecting up to 60% of all cancer patients receiving chemotherapy. Moreover, paclitaxel induces neuropathy in up to 97% of all gynecological and urological cancer patients. In cancer cells, paclitaxel induces cell death via microtubule stabilization interrupting cell mitosis. However, paclitaxel also affects cells of the central and peripheral nervous system. The main symptoms are pain and numbness in hands and feet due to paclitaxel accumulation in the dorsal root ganglia. This review describes in detail the pathomechanisms of paclitaxel in the peripheral nervous system. Symptoms occur due to a length-dependent axonal sensory neuropathy, where axons are symmetrically damaged and die back. Due to microtubule stabilization, axonal transport is disrupted, leading to ATP undersupply and oxidative stress. Moreover, mitochondria morphology is altered during paclitaxel treatment. A key player in pain sensation and axonal damage is the paclitaxel-induced inflammation in the spinal cord as well as the dorsal root ganglia. An increased expression of chemokines and cytokines such as IL-1ß, IL-8, and TNF-α, but also CXCR4, RAGE, CXCL1, CXCL12, CX3CL1, and C3 promote glial activation and accumulation, and pain sensation. These findings are further elucidated in this review.

12.
J Neurol Sci ; 427: 117507, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34130060

ABSTRACT

Chronic inflammatory demyelinating polyneuropathy (CIDP) is an autoimmune disorder causing inflammatory demyelination of peripheral nerves and consecutive disability. Diagnostic criteria and treatments are well established, but it is unknown how clinical practice may differ in different geographical regions. In this multicentre study, clinical management of CIDP was compared in 44 patients from Germany, India and Norway regarding diagnostic and therapeutic procedures. All centres used EFNS/PNS diagnostic criteria for CIDP but diagnostic workup varied regarding screening for infectious diseases, genetic testing and nerve biopsy. Intravenous immunoglobulin and prednisolone were the most common therapies in all centres with differences in indication and dosage. Patients from the Indian cohort were the most severely affected with less diverse therapeutic approaches, whereas psychological strain did not differ significantly from the two other cohorts. Our exploratory study discloses an unaddressed issue in management of CIDP that should be further investigated to optimise standard of care for CIDP worldwide.


Subject(s)
Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Biopsy , Europe , Humans , Immunoglobulins, Intravenous/therapeutic use , Peripheral Nerves , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/diagnosis , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/epidemiology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/therapy
13.
J Peripher Nerv Syst ; 26(2): 216-226, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33683765

ABSTRACT

Peripheral neuropathy is a common side effect of paclitaxel. Clinical studies suggest that different paclitaxel formulations influence the severity and time course of paclitaxel-induced peripheral neuropathy. We compared two paclitaxel formulations, nanoparticle albumin-bound paclitaxel (nab-paclitaxel) and Cremophor EL paclitaxel (CreEL-paclitaxel), for their toxicity, distribution, and clearance in the peripheral nervous system. Neuronal F11 cells were used to detect changes in morphology, cell nuclei size, and cell viability after nab- or CreEL-paclitaxel treatment via MTT Assay and immunohistochemistry. C57BL/6 mice were treated with 50 mg/kg of nab-paclitaxel or CreEL-paclitaxel. Paclitaxel levels in serum, liver, dorsal root ganglia (DRG), and sciatic nerve (SCN) were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Accumulation of paclitaxel in DRG neurons and SCN was visualized by immunostainings. Neurotoxicity was evaluated after a 4-week treatment regime with nab- or CreEL-paclitaxel by nerve morphology, behavioral, and functional assays. In vitro cell nuclei size and morphology were similar between the two treatment groups. Viability was increased in neurons exposed to nab-paclitaxel compared to CreEL-paclitaxel. In vivo paclitaxel mostly accumulated in DRG. SCN displayed lower paclitaxel uptake. The two paclitaxel formulations mainly accumulated in neurofilament 200-positive large-caliber neurons and less in Isolectin B4-, or calcitonin gene-related peptide-positive small-caliber neurons. Sensory nerve conduction studies demonstrated increased sensory latencies after 11 days in nab-paclitaxel treated animals, while an increase occurred after 22 days in CreEL-paclitaxel treated animals. Behavioral testing did not reveal significant differences between the different groups. Skin denervation, axon count, myelin thickness, and F4/80-positive cell accumulation were comparable between the two treatment groups. Our findings indicate that different drug formulations impact the severity of neuropathy induced by paclitaxel via different tissue uptake. Neurotoxicity was comparable between the two paclitaxel formulations.


Subject(s)
Neurotoxicity Syndromes , Peripheral Nervous System Diseases , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Agents, Phytogenic/toxicity , Chromatography, Liquid , Drug Compounding , Ganglia, Spinal , Kinetics , Mice , Mice, Inbred C57BL , Paclitaxel/toxicity , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Tandem Mass Spectrometry
14.
J Neuroimmunol ; 352: 577470, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33508768

ABSTRACT

Nerve conduction studies (NCS) are essential to assess peripheral nerve fiber function in research models of immune-mediated neuritis. However, the current lack of standard protocols and reference values impedes data comparability across models and studies. We performed a systematic review and subsequent meta-analysis of the last 30 years of NCS of immune-mediated neuritis in Lewis-rats. Twenty-six papers met the inclusion criteria for meta-analysis. Extracted data showed considerable heterogeneity of recorded nerve conduction velocity (NCV) and compound muscle action potential (CMAP). Studies also significantly differed in terms of technical, methodical, and data reporting issues. The heterogeneity of the underlying studies emphasizes the need for standardization when conducting and reporting NCS in rats. We provide normative values for NCS of the sciatic nerve of Lewis rats and propose seven items that should be addressed when NCS are performed when studying immune paradigms in Lewis rats.


Subject(s)
Electrophysiology/methods , Electrophysiology/standards , Neuritis, Autoimmune, Experimental/physiopathology , Animals , Neural Conduction/physiology , Rats, Inbred Lew , Reference Values , Sciatic Nerve/physiology
15.
PLoS One ; 12(10): e0178372, 2017.
Article in English | MEDLINE | ID: mdl-29040264

ABSTRACT

BACKGROUND: Transpulmonary thermodilution (TPTD) is used to derive cardiac output CO, global end-diastolic volume GEDV and extravascular lung water EVLW. To facilitate interpretation of these data, several ratios have been developed, including pulmonary vascular permeability index (defined as EVLW/(0.25*GEDV)) and global ejection fraction ((4*stroke volume)/GEDV). PVPI and GEF have been associated to the aetiology of pulmonary oedema and systolic cardiac function, respectively. Several studies demonstrated that the use of femoral venous access results in a marked overestimation of GEDV. This also falsely reduces PVPI and GEF. One of these studies suggested a correction formula for femoral venous access that markedly reduced the bias for GEDV. Consequently, the last PiCCO-algorithm requires information about the CVC, and correction for femoral access has been shown. However, two recent studies demonstrated inconsistencies of the last PiCCO algorithm using incorrected GEDV for PVPI, but corrected GEDV for GEF. Nevertheless, these studies were based on mathematical analyses of data displayed in a total of 15 patients equipped with only a femoral, but not with a jugular CVC. Therefore, this study compared PVPI_fem and GEF_fem derived from femoral TPTD to values derived from jugular indicator injection in 25 patients with both jugular and femoral CVCs. METHODS: 54 datasets in 25 patients were recorded. Each dataset consisted of three triplicate TPTDs using the jugular venous access as the gold standard and the femoral access with (PVPI_fem_cor) and without (PVPI_fem_uncor) information about the femoral indicator injection to evaluate, if correction for femoral GEDV pertains to PVPI_fem and GEF_fem. RESULTS: PVPI_fem_uncor was significantly lower than PVPI_jug (1.48±0.47 vs. 1.84±0.53; p<0.001). Similarly, PVPI_fem_cor was significantly lower than PVPI_jug (1.49±0.46 vs. 1.84±0.53; p<0.001). This is explained by the finding that PVPI_fem_uncor was not different to PVPI_fem_cor (1.48±0.47 vs. 1.49±0.46; n.s.). This clearly suggests that correction for femoral CVC does not pertain to PVPI. GEF_fem_uncor was significantly lower than GEF_jug (20.6±5.1% vs. 25.0±6.1%; p<0.001). By contrast, GEF_fem_cor was not different to GEF_jug (25.6±5.8% vs. 25.0±6.1%; n.s.). Furthermore, GEF_fem_cor was significantly higher than GEF_fem_uncor (25.6±5.8% vs. 20.6±5.1%; p<0.001). This finding emphasizes that an appropriate correction for femoral CVC is applied to GEF_fem_cor. The extent of the correction (25.5/20.6; 124%) for GEF and the relation of PVPI_jug/PVPI_fem_uncor (1.84/1.48; 124%) are in the same range as the ratio of GEDVI_fem_uncor/GEDVI_fem_cor (1056ml/m2/821mL/m2; 129%). This further emphasizes that GEF, but not PVPI is corrected in case of femoral indicator injection. CONCLUSIONS: Femoral indicator injection for TPTD results in significantly lower values for PVPI and GEF. While the last PiCCO algorithm appropriately corrects GEF, the correction is not applied to PVPI. Therefore, GEF-values can be used in case of femoral CVC, but PVPI-values are substantially underestimated.


Subject(s)
Capillary Permeability , Cardiac Output , Catheterization, Central Venous/methods , Contrast Media/pharmacokinetics , Monitoring, Physiologic/methods , Stroke Volume , Aged , Extravascular Lung Water , Female , Femoral Vein , Heart/physiopathology , Humans , Injections, Intravenous , Intensive Care Units , Jugular Veins , Liver Cirrhosis/diagnosis , Liver Cirrhosis/physiopathology , Liver Cirrhosis/therapy , Lung/blood supply , Lung/physiopathology , Middle Aged , Monitoring, Physiologic/instrumentation , Prospective Studies , Pulmonary Edema/diagnosis , Pulmonary Edema/physiopathology , Pulmonary Edema/therapy , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/physiopathology , Respiratory Distress Syndrome/therapy , Sepsis/diagnosis , Sepsis/physiopathology , Sepsis/therapy , Thermodilution/methods
16.
Neurotox Res ; 32(4): 555-562, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28612296

ABSTRACT

Neurotoxicity is a relevant side effect of bortezomib treatment. Previous reports have shown that the development of peripheral neuropathy caused by anti-neoplastic agents may be a result of reduced axonal transport. Based on evidence from prior studies that the kinesin-5 inhibitor monastrol enhances axonal transport and improves neuronal regeneration, we focused on the neuroprotective role of monastrol during the chemotherapeutic treatment with bortezomib. Prolonged treatment of C57BL/6 mice with bortezomib induced a length-dependent small-fiber neuropathy with axonal atrophy and loss of sensory nerve fibers. The administration of monastrol substantially alleviated morphological features of axonal injury and functional measures of sensory neuropathy. Cytotoxicity studies in leukemia and multiple myeloma cell lines showed no interference of monastrol with the cytostatic effects of bortezomib. Our data indicate that the novel approach of targeting microtubule turnover by monastrol provides protection against bortezomib-induced neurotoxicity. The favorable cytotoxic profile of monastrol makes it an interesting candidate as neuroprotective agent in combined chemotherapy regimens that warrants further consideration.


Subject(s)
Kinesins/antagonists & inhibitors , Neurons/drug effects , Neurotoxicity Syndromes/drug therapy , Pyrimidines/pharmacology , Thiones/pharmacology , Animals , Antineoplastic Agents/pharmacology , Axons/drug effects , Bortezomib/pharmacology , Cells, Cultured , Mice, Inbred C57BL , Microtubules/drug effects , Microtubules/metabolism , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Peripheral Nervous System/drug effects
17.
Glia ; 63(6): 977-86, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25628221

ABSTRACT

We investigated connexin 32 (Cx32)-deficient mice, a model for the X-linked form of Charcot-Marie-Tooth neuropathy (CMT1X), regarding the impact of low-grade inflammation on Schwann cell phenotype. Whereas we previously identified macrophages as amplifiers of the neuropathy, we now explicitly focus on the impact of the phagocytes on Schwann cell dedifferentiation, a so far not-yet addressed disease-related mechanism for CMT1X. Using mice heterozygously deficient for Cx32 and displaying both Cx32-positive and -negative Schwann cells in one and the same nerve, we could demonstrate that macrophage clusters rather than single macrophages precisely associate with mutant but not with Cx32-positive Schwann cells. Similarly, in an advanced stage of Schwann cell perturbation, macrophage clusters were strongly associated with NCAM- and L1-positive, dedifferentiated Schwann cells. To clarify the role of macrophages regarding Schwann cell dedifferentiation, we generated Cx32-deficient mice additionally deficient for the macrophage-directed cytokine colony-stimulating factor (CSF)-1. In the absence of CSF-1, Cx32-deficient Schwann cells not only showed the expected amelioration in myelin preservation but also failed to upregulate the Schwann cell dedifferentiation markers NCAM and L1. Another novel and unexpected finding in the double mutants was the retained activation of ERK signaling, a pathway which is detrimental for Schwann cell homeostasis in myelin mutant models. Our findings demonstrate that increased ERK signaling can be compatible with the maintenance of Schwann cell differentiation and homeostasis in vivo and identifies CSF-1-activated macrophages as crucial mediators of detrimental Schwann cell dedifferentiation in Cx32-deficient mice.


Subject(s)
Connexins/deficiency , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/physiology , Schwann Cells/physiology , Animals , Cell Dedifferentiation/physiology , Charcot-Marie-Tooth Disease , Chemokine CCL2/metabolism , Connexins/genetics , Disease Models, Animal , Female , Homeostasis/physiology , MAP Kinase Signaling System/physiology , Macrophage Colony-Stimulating Factor/genetics , Macrophages/ultrastructure , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/metabolism , Neural Cell Adhesion Molecules/metabolism , Schwann Cells/ultrastructure , Sciatic Nerve/physiopathology , Sciatic Nerve/ultrastructure , Gap Junction beta-1 Protein
18.
Eur J Pediatr Surg ; 25(2): 216-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24623454

ABSTRACT

BACKGROUND: Prospective data of transumbilically laparoscopic-assisted appendectomies (TULAA) is absent in the pediatric population. We therefore compared the midterm outcome of TULAA with open (OA) and laparoscopic (LA) appendectomies in children with appendicitis in a matched prospective study. METHODS: A total of 20 patients operated with TULAA were matched to 20 cases operated by LA and OA, respectively, according to sex, age, and histology of the resected appendix. All 60 children were evaluated during a 3-month follow-up visit. RESULTS: The subjective pain level after discharge, the rate of complications, and persistent painful wound as well as the duration of days refraining from school or kindergarten were similar in all three groups. The wound satisfaction was significantly higher in TULAA and LA. Children operated with TULAA had a faster return to full physical activity compared with OA. CONCLUSION: Our data suggest that TULAA, LA, and OA have a similar outcome 3 months after surgery apart from cosmetic appearance of the wound and return to full physical activity in pediatric patients. Whether parental bias or the increased wound satisfaction act as confounders for early return to full physical activity should be evaluated in larger prospective randomized trials.


Subject(s)
Appendectomy/methods , Appendicitis/surgery , Laparoscopy/methods , Adolescent , Appendectomy/adverse effects , Child , Female , Humans , Laparoscopy/adverse effects , Length of Stay , Male , Pain, Postoperative , Pilot Projects , Prospective Studies , Treatment Outcome , Umbilicus/surgery , Wound Healing
19.
Surg Laparosc Endosc Percutan Tech ; 24(3): 244-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24710220

ABSTRACT

INTRODUCTION: Aim of this study was to compare the short-term outcome of transumbilical laparoscopic-assisted (TULAA) with laparoscopic (LA) and open appendectomy (OA) in a case-control study. MATERIALS AND METHODS: Our first 20 consecutive children with appendicitis treated with TULAA were matched to 20 patients treated with LA and OA, respectively. Matching criteria were age, sex, and the histology of appendicitis. The children were retrospectively evaluated for outcome, efficacy, and complications. RESULTS: No significant differences between the groups were found, except that children operated by OA required less analgesics than children operated by LA and TULAA. DISCUSSION: TULAA appears to be a safe procedure with no disadvantage except for postoperative pain, which needed a longer course of analgesics compared to OA. Whether TULAA has advantages over LA and OA has to be evaluated in larger prospective series.


Subject(s)
Appendectomy/methods , Appendicitis/surgery , Laparoscopes , Laparoscopy/methods , Laparotomy/methods , Acute Disease , Adolescent , Child , Equipment Design , Female , Germany/epidemiology , Humans , Incidence , Length of Stay/trends , Male , Pain, Postoperative/epidemiology , Retrospective Studies , Umbilicus
SELECTION OF CITATIONS
SEARCH DETAIL
...