Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Nat Neurosci ; 27(5): 901-912, 2024 May.
Article En | MEDLINE | ID: mdl-38514857

Multiple sclerosis (MS) is an autoimmune disease characterized by demyelination of the central nervous system (CNS). Autologous hematopoietic cell transplantation (HCT) shows promising benefits for relapsing-remitting MS in open-label clinical studies, but the cellular mechanisms underlying its therapeutic effects remain unclear. Using single-nucleus RNA sequencing, we identify a reactive myeloid cell state in chronic experimental autoimmune encephalitis (EAE) associated with neuroprotection and immune suppression. HCT in EAE mice results in an increase of the neuroprotective myeloid state, improvement of neurological deficits, reduced number of demyelinated lesions, decreased number of effector T cells and amelioration of reactive astrogliosis. Enhancing myeloid cell incorporation after a modified HCT further improved these neuroprotective effects. These data suggest that myeloid cell manipulation or replacement may be an effective therapeutic strategy for chronic inflammatory conditions of the CNS.


Encephalomyelitis, Autoimmune, Experimental , Mice, Inbred C57BL , Myeloid Cells , Animals , Encephalomyelitis, Autoimmune, Experimental/therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Female , Hematopoietic Stem Cell Transplantation/methods , Neuroprotection/physiology
2.
bioRxiv ; 2023 Mar 12.
Article En | MEDLINE | ID: mdl-36945385

Multiple sclerosis (MS) is an autoimmune disease associated with inflammatory demyelination in the central nervous system (CNS). Autologous hematopoietic cell transplantation (HCT) is under investigation as a promising therapy for treatment-refractory MS. Here we identify a reactive myeloid state in chronic experimental autoimmune encephalitis (EAE) mice and MS patients that is surprisingly associated with neuroprotection and immune suppression. HCT in EAE mice leads to an enhancement of this myeloid state, as well as clinical improvement, reduction of demyelinated lesions, suppression of cytotoxic T cells, and amelioration of reactive astrogliosis reflected in reduced expression of EAE-associated gene signatures in oligodendrocytes and astrocytes. Further enhancement of myeloid cell incorporation into the CNS following a modified HCT protocol results in an even more consistent therapeutic effect corroborated by additional amplification of HCT-induced transcriptional changes, underlining myeloid-derived beneficial effects in the chronic phase of EAE. Replacement or manipulation of CNS myeloid cells thus represents an intriguing therapeutic direction for inflammatory demyelinating disease.

3.
Exp Ther Med ; 20(2): 1455-1466, 2020 Aug.
Article En | MEDLINE | ID: mdl-32742379

Carboxyamidotriazole (CAI), originally developed as a non-cytotoxic anti-cancer drug, was shown to have anti-inflammatory activity according to recent studies in a number of animal models of inflammation. However, its mechanism of action has not been characterized. Therefore, the present study was performed to identify the anti-inflammatory action of CAI in lipopolysaccharide (LPS)-induced RAW 264.7 macrophages and to identify the signal transduction pathways involved. The in vitro results revealed that CAI had no direct effect on the activity of cyclooxygenase (COX), suggesting a different anti-inflammatory mechanism compared with that of COX-inhibiting non-steroidal anti-inflammatory drugs. Further investigation in RAW264.7 macrophages revealed that CAI decreased the production of nitric oxide via decreasing the LPS-stimulated expression of inducible nitric oxide synthase, and downregulated both mRNA and protein expression levels of the cytokines tumor necrosis factor-α, interleukin (IL)-1ß, and IL-6. CAI also significantly reduced the increased DNA-binding activity of nuclear factor (NF)-κB induced by LPS stimulation. With respect to the mechanisms involved on NF-κB activity, CAI exhibited suppression of the phosphorylation and degradation of the inhibitor of nuclear factor-κBα (IκB), and decreased the phosphorylation levels of the p65 subunit and its subsequent nuclear translocation. In addition, CAI significantly decreased the phosphorylated forms of p38, JNK and ERK, which were increased following LPS stimulation, while the total expression levels of p38, JNK and ERK remained unaltered. The results in the present study indicate that CAI alleviates the inflammatory responses of RAW 264.7 macrophages in response to LPS stimulation via attenuating the activation of NF-κB and MAPK signaling pathways and decreasing the levels of pro-inflammatory mediators. This offers a novel perspective for understanding the anti-inflammatory mechanism of CAI and suggests its potential use as a therapeutic treatment in inflammatory diseases with excessive macrophage activation.

4.
Proc Natl Acad Sci U S A ; 116(30): 15262-15271, 2019 07 23.
Article En | MEDLINE | ID: mdl-31285321

Giant ankyrin-B (ankB) is a neurospecific alternatively spliced variant of ANK2, a high-confidence autism spectrum disorder (ASD) gene. We report that a mouse model for human ASD mutation of giant ankB exhibits increased axonal branching in cultured neurons with ectopic CNS axon connectivity, as well as with a transient increase in excitatory synapses during postnatal development. We elucidate a mechanism normally limiting axon branching, whereby giant ankB localizes to periodic axonal plasma membrane domains through L1 cell-adhesion molecule protein, where it couples microtubules to the plasma membrane and prevents microtubule entry into nascent axon branches. Giant ankB mutation or deficiency results in a dominantly inherited impairment in selected communicative and social behaviors combined with superior executive function. Thus, gain of axon branching due to giant ankB-deficiency/mutation is a candidate cellular mechanism to explain aberrant structural connectivity and penetrant behavioral consequences in mice as well as humans bearing ASD-related ANK2 mutations.


Ankyrins/genetics , Autism Spectrum Disorder/genetics , Neural Cell Adhesion Molecule L1/genetics , Neuronal Outgrowth , Neurons/metabolism , Synapses/metabolism , Alternative Splicing , Animals , Ankyrins/metabolism , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/physiopathology , Behavior, Animal , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Connectome , Disease Models, Animal , Executive Function/physiology , Gene Expression , Gene Knock-In Techniques , Humans , Male , Mice , Mice, Transgenic , Microtubules/metabolism , Microtubules/ultrastructure , Mutation , Neural Cell Adhesion Molecule L1/metabolism , Neurons/pathology , Primary Cell Culture , Social Behavior , Synapses/pathology
5.
Cureus ; 10(8): e3202, 2018 Aug 24.
Article En | MEDLINE | ID: mdl-30405981

We report a case of a 61-year-old man with a history of squamous cell carcinoma of the lung presenting with rapidly progressive symmetric ascending weakness with areflexia. The weakness was quickly followed by respiratory decompensation requiring intubation. Lumbar puncture yielded cerebrospinal fluid with elevated protein (177 mg/dL), normal glucose (61 mg/dL), normal red blood cell count (0 per/µl), and normal white blood cell count (0 per/µL). Emergent magnetic resonance imaging of cervical, thoracic, and lumbar spine did not show evidence of metastatic disease, fracture, subluxation, or other causes of cord compression. The patient was diagnosed with acute inflammatory polyneuropathy, also known as Guillain-Barré syndrome. Despite treatment with a five-day course of intravenous immunoglobulin and a subsequent five-day course of plasmapheresis, the patient did not recover respiratory function and died 48 days after diagnosis. To our knowledge, this is the first documented case of Guillain-Barré occurring concomitantly with squamous cell carcinoma of the lung.

6.
Cureus ; 10(7): e3061, 2018 Jul 28.
Article En | MEDLINE | ID: mdl-30397566

Neonatal abstinence syndrome (NAS) is a neurologic condition resulting from prenatal exposure to opioids. The sudden cessation of opioids in neonates can lead to withdrawal symptoms affecting the neurologic, respiratory, and gastrointestinal systems. Rising opioid use in the United States has led to an increased incidence of infants born with NAS. Despite the growing incidence of NAS, there is a lack of standardized guidelines for intervention and management. Recent studies suggest that non-pharmacological methods should be used as first-line interventions for the reduction of NAS symptoms. Of the non-pharmacological methods, growing literature suggests that breastfeeding may have the potential to reduce symptom severity and improve outcomes. We searched the PubMed and Medline databases for experimental/quasi-experimental studies published from 1997-2018 regarding outcomes in breastfed versus formula-fed neonates with prenatal exposure to opioids. Seven retrospective studies fulfilling the inclusion criteria were reviewed. Collectively, the studies show a strong correlation between breastfeeding and a reduced length of hospital stay, a decreased severity of NAS presentation, and a decreased necessity of pharmacological interventions in infants diagnosed with NAS. From these findings, we recommend breastfeeding as an integral component of the early management of NAS.

7.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 38(1): 49-54, 2016 Feb.
Article En | MEDLINE | ID: mdl-26956856

OBJECTIVE: To study the effect of carboxyamidotriazole (CAI) on adjuvant arthritis (AA) in rats. METHODS: The rats were randomly divided into normal group,two vehicle groups (polyethylene glycol 400 control and normal sodium control group), CAI-treated groups (10, 20, and 40 mg/kg) and positive control dexamethasone group. Freund's completed adjuvant was used to induce AA in rats. The arthritis index (AI) was scored, and X-ray check of the hind limbs and histopathological examination were performed. The levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 in the inflamed paw tissues were measured. RESULTS: The administration of CAI significantly decreased the AI, restored the body weights, and ameliorated the radiological and histopathological features of joint destruction in AA rats (P<0.05, P<0.01). In addition, CAI reduced the TNF-α, IL-1ß, and IL-6 levels in the inflamed paw tissues (P<0.05, P<0.01). CONCLUSION: CAI has therapeutic effect on AA rats, which may be achieved by decreasing the pro-inflammatory cytokines at the site of inflammation.


Arthritis, Experimental , Animals , Freund's Adjuvant , Interleukin-1beta , Interleukin-6 , Rats , Triazoles , Tumor Necrosis Factor-alpha
8.
J Neurosci ; 35(18): 7003-18, 2015 May 06.
Article En | MEDLINE | ID: mdl-25948253

The cerebral cortex is built during embryonic neurogenesis, a period when excitatory neurons are generated from progenitors. Defects in neurogenesis can cause acute neurodevelopmental disorders, such as microcephaly (reduced brain size). Altered dosage of the 1q21.1 locus has been implicated in the etiology of neurodevelopmental phenotypes; however, the role of 1q21.1 genes in neurogenesis has remained elusive. Here, we show that haploinsufficiency for Rbm8a, an exon junction complex (EJC) component within 1q21.1, causes severe microcephaly and defective neurogenesis in the mouse. At the onset of neurogenesis, Rbm8a regulates radial glia proliferation and prevents premature neuronal differentiation. Reduced Rbm8a levels result in subsequent apoptosis of neurons, and to a lesser extent, radial glia. Hence, compared to control, Rbm8a-haploinsufficient brains have fewer progenitors and neurons, resulting in defective cortical lamination. To determine whether reciprocal dosage change of Rbm8a alters embryonic neurogenesis, we overexpressed human RBM8A in two animal models. Using in utero electroporation of mouse neocortices as well as zebrafish models, we find RBM8A overexpression does not significantly perturb progenitor number or head size. Our findings demonstrate that Rbm8a is an essential neurogenesis regulator, and add to a growing literature highlighting roles for EJC components in cortical development and neurodevelopmental pathology. Our results indicate that disruption of RBM8A may contribute to neurodevelopmental phenotypes associated with proximal 1q21.1 microdeletions.


Cerebral Cortex/embryology , Embryonic Development/physiology , Haploinsufficiency/physiology , Microcephaly/genetics , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microcephaly/metabolism , Organogenesis/physiology
9.
Br J Pharmacol ; 172(13): 3446-59, 2015 Jul.
Article En | MEDLINE | ID: mdl-25799914

BACKGROUND AND PURPOSE: Pro-inflammatory cytokines are important in rheumatoid arthritis (RA) and their production is mainly regulated by NF-κB and inflammasomes. Carboxyamidotriazole (CAI) exhibits potent anti-inflammatory activities by decreasing cytokines. Here, we have investigated NACHT, LRR and PYD domains-containing protein (NALP) inflammasomes in a rat model of RA and explored the therapeutic effects of CAI in this model and the involvement of NF-κB and inflammasomes in the actions of CAI. EXPERIMENTAL APPROACH: The anti-arthritic effects of CAI were assessed in the adjuvant arthritis (AA) model in rats, using radiological and histological techniques. NALP1 and NALP3 inflammasomes, NF-κB pathway and pro-inflammatory cytokines levels were measured with Western blots, immunohistochemistry and ELISA. KEY RESULTS: CAI decreased the arthritis index, improved radiological and histological changes, and reduced synovial IL-1ß, IL-6, IL-18 and TNF-α levels in rats with AA. Compared with normal rats, the 70 kDa NALP1 isoform was up-regulated, NALP3 was down-regulated, and levels of the 165 kDa NALP1 isoform and the adaptor protein ASC were unchanged in synovial tissue from AA rats. CAI reduced the 70 kDa NALP1 isoform and restored NALP3 levels in AA rats; CAI inhibited caspase-1 activation in AA synovial tissue, but not its enzymic activity in vitro. In addition, CAI reduced expression of p65 NF-κB subunit and IκBα phosphorylation and degradation in AA rats. CONCLUSION AND IMPLICATIONS: NALP1 inflammasomes were activated in synovial tissues from AA rats and appeared to be a novel therapeutic target for RA. CAI could have therapeutic value in RA by inhibiting activation of NF-κB and NALP1 inflammasomes and by decreasing pro-inflammatory cytokines.


Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Inflammasomes/metabolism , Nerve Tissue Proteins/metabolism , Triazoles/pharmacology , Animals , Ankle Joint/diagnostic imaging , Ankle Joint/drug effects , Ankle Joint/metabolism , Ankle Joint/pathology , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/diagnostic imaging , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/diagnostic imaging , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Cytokines/metabolism , Male , Radiography , Rats, Inbred Lew , Synovial Membrane/metabolism , Triazoles/therapeutic use
10.
J Pharmacol Exp Ther ; 342(2): 356-65, 2012 Aug.
Article En | MEDLINE | ID: mdl-22553216

Carboxyamidotrizole (CAI) has been reported to suppress the production of tumor necrosis factor-α (TNF-α) and interleukin (IL)-1ß and be effective in rats with adjuvant arthritis. The aim of this study was to investigate the role of CAI in inflammatory bowel disease (IBD). We assessed the effect of CAI in dextran sodium sulfate-induced colitis. Inflammation was scored histologically, and potential mediators of IBD were assessed by immunohistochemical and molecular biochemical approaches. CAI-treated colitis animals revealed much fewer signs of colitis with significantly decreased macroscopic and microscopic scores than vehicle-treated animals. CAI inhibited the production of TNF-α, IL-1ß, and IL-6 in serum, supernatant of peritoneal macrophages, and lamina propria. CAI also decreased the expression of intercellular adhesion molecule-1 in colonic tissues. Furthermore, CAI prevented nuclear factor-κB (NF-κB) activation and inhibitor of nuclear factor-κBα phosphorylation and degradation. In addition, CAI showed a beneficial effect on colonic fibrosis, possibly by reducing the production of the fibrogenic cytokine transforming growth factor-ß. The results support that CAI administration is effective in ameliorating experimental colitis and preventing colonic fibrosis. The inhibition of proinflammatory cytokines and adhesion molecules and suppression of NF-κB activation seem to contribute to this effect.


Colitis/drug therapy , Colon/drug effects , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Triazoles/pharmacology , Animals , Colitis/blood , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Dextran Sulfate , Fibrosis , Inflammation/blood , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammatory Bowel Diseases/blood , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1beta/blood , Interleukin-1beta/metabolism , Interleukin-6/blood , Interleukin-6/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mucous Membrane/drug effects , Mucous Membrane/metabolism , Mucous Membrane/pathology , Phosphorylation/drug effects , Proteolysis/drug effects , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
11.
Eur J Cancer ; 48(7): 1085-95, 2012 May.
Article En | MEDLINE | ID: mdl-21767946

Carboxyamidotriazole (CAI) has not only direct anti-cancer activities, but also anti-inflammation effects in a variety of inflammatory animal models. In the present study, we investigated whether macrophages, which are important both in cancer and inflammation, could be regulated by CAI. The results showed that CAI could inhibit tumour necrosis factor-α (TNF-α) production in macrophages in various environments, including those isolated from peritoneal cavity of adjuvant-induced arthritis (AA) rats, from Lewis lung carcinoma (LLC) transplanted tumours and those induced by LLC cells in vitro. Dexamethasone (DEX), one of the pro-inflammatory cytokines inhibitors, could enhance CAI's inhibition of LLC cells proliferation and invasion in macrophages and LLC cells co-culture systems, as well as the tumour growth in vivo. However, DEX failed to enhance CAI's inhibition of LLC cells proliferation when LLC cells were cultured alone, suggesting that the combination of CAI and DEX exerted great anti-tumour effects probably by acting on macrophages in the tumour environment. Over all, we found CAI could act on macrophages and regulate the production of TNF-α not only in inflammatory diseases but also in tumour microenvironment, which might be another anti-tumour mechanism of CAI.


Anti-Inflammatory Agents/pharmacology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/metabolism , Macrophages/drug effects , Neoplasms/drug therapy , Triazoles/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/blood , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Coculture Techniques , Cytokines , Dexamethasone/pharmacology , Macrophages/immunology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms/immunology , Rats, Wistar , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
12.
J Electron Microsc (Tokyo) ; 58(1): 15-9, 2009 Jan.
Article En | MEDLINE | ID: mdl-19029106

We clarify the scanning electron microscopic contrast mechanism for imaging a buried SiO(2) trench microstructure with interface trapped charges by simulating both electron scattering and transport. Here, the interface trapped charges make the SiO(2) film more negatively charged and increase excess holes in the space charge distribution of the electron scattering region. The generated positive surface electric field thus redistributes some emitted secondary electrons and results in the dark contrast. This contrast mechanism is validated by comparing with experiments, and it may also provide an interesting approach for imaging and detecting deep interface trapped charges in insulating films.

...