Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Biochem Pharmacol ; 228: 116302, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38763261

ABSTRACT

Spinal cord injury (SCI) afflicts millions of individuals globally. There are few therapies available to patients. Ascending and descending excitatory glutamatergic neural circuits in the central nervous system are disrupted by SCI, making α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) a potential therapeutic drug target. Emerging research in preclinical models highlights the involvement of AMPARs in vital processes following SCI including breathing, pain, inflammation, bladder control, and motor function. However, there are no clinical trial data reported in this patient population to date. No work on the role of AMPA receptors in sexual dysfunction after SCI has been disclosed. Compounds with selective antagonist and potentiating effects on AMPA receptors have benefit in animal models of SCI, with antagonists generally showing protective effects early after injury and potentiators (ampakines) producing improved breathing and bladder function. The role of AMPARs in pathophysiology and recovery after SCI depends upon the time post injury, and the timing of AMPAR augmentation or antagonism. The roles of inflammation, synaptic plasticity, sensitization, neurotrophic factors, and neuroprotection are considered in this context. The data summarized and discussed in this paper document proof of principle and strongly encourage additional studies on AMPARs as novel gateways to therapeutic benefit for patients suffering from SCI. The availability of both AMPAR antagonists such as perampanel and AMPAR allosteric modulators (i.e., ampakines) such as CX1739, that have been safely administered to humans, provides an expedited means of clinical inquiry for possible therapeutic advances.


Subject(s)
Receptors, AMPA , Spinal Cord Injuries , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism , Animals , Humans , Excitatory Amino Acid Antagonists/therapeutic use , Excitatory Amino Acid Antagonists/pharmacology
2.
Clin Transl Discov ; 2(4)2022 Dec.
Article in English | MEDLINE | ID: mdl-36268259

ABSTRACT

Introduction: The aim of this review is to summarize the current understanding of the gut-brain axis (GBA), its impact on neurosurgery, and its implications for future treatment. Background: An abundance of research has established the existence of a collection of pathways between the gut microbiome and the central nervous system (CNS), commonly known as the GBA. Complicating this relationship, the gut microbiome bacterial diversity appears to change with age, antibiotic exposure and a number of external and internal factors. Methods: In this paper, we present the current understanding of the key protective and deleterious roles the gut microbiome plays in the pathogenesis of several common neurosurgical concerns. Results: Specifically, we examine how spinal cord injury, traumatic brain injury and stroke may cause gut microbial dysbiosis. Furthermore, this link appears to be bidirectional as gut dysbiosis contributes to secondary CNS injury in each of these ailment settings. This toxic cycle may be broken, and the future secondary damage rescued by timely, therapeutic, gut microbiome modification. In addition, a robust gut microbiome appears to improve outcomes in brain tumour treatment. There are several primary routes by which microbiome dysbiosis may be ameliorated, including faecal microbiota transplant, oral probiotics, bacteriophages, genetic modification of gut microbiota and vagus nerve stimulation. Conclusion: The GBA represents an important component of patient care in the field of neurosurgery. Future research may illuminate ideal methods of therapeutic microbiome modulation in distinct pathogenic settings.

3.
AIMS Allergy Immunol ; 6(4): 216-227, 2022.
Article in English | MEDLINE | ID: mdl-36285334

ABSTRACT

Contrast-enhanced neuroimaging is often necessary for the diagnosis and care of patients with diseases of the central nervous system. Although contrast is generally well tolerated and allergy to contrast is rare, allergic reactions can be severe and life threatening. Therefore, physicians should take care to prevent severe contrast allergy. In this review, we will discuss contrast allergy as well as potential strategies to reduce the risk of severe reactions in patients who require neuroimaging techniques with contrast. First, we discuss the clinical presentation and pathogenesis of contrast allergy and the risk factors associated with reactions. We then review methods to reduce the risk of future contrast reactions through improved patient education and documentation strategies, use of alternate imaging modalities or contrast media, premedication, and desensitization.

4.
Trauma Emerg Med ; 1(1): 1-12, 2022.
Article in English | MEDLINE | ID: mdl-36745142

ABSTRACT

Focused ultrasound has emerged as a key tool for neurologic disorders. In this focused review, we discuss the utility in disrupting the blood brain barrier to maximize treatment. This can facilitate creating direct coagulative lesions and aid in the administration of chemotherapy. Furthermore, it can facilitate neuromodulation when used in pulse sequencing. The current literature regarding brain tumors, essential tremor, and obsessive-compulsive disorder is reviewed. Additionally, concepts and experimental outcomes for neurodegenerative disease such as Alzheimer's is presented. Focused ultrasound as a tool is still in its infancy but the potential for adjuvant and direct therapy is promising. More clinical uses will become apparent in coming decades.

5.
Article in English | MEDLINE | ID: mdl-36745148

ABSTRACT

Chronic traumatic encephalopathy (CTE) is a neurodegenerative disease associated with repeated head injury. The common presenting neuropsychiatric manifestations and diagnostic strategies for early diagnosis and subsequent treatment will be reviewed. This article discusses methods for injury prevention, risk assessment, and methods for supportive symptom management including lifestyle modifications, physical, occupational, and neurorehabilitation, and pharmaceutical management. Lastly, we propose the use of assessment tools validated for other neurodegenerative disorders in CTE to establish a baseline, track outcomes, and measure improvement in this population.

6.
Explor Neurosci ; 1(2): 83-99, 2022.
Article in English | MEDLINE | ID: mdl-36655054

ABSTRACT

Stroke is a leading cause of morbidity and mortality. The advent of mechanical thrombectomy has largely improved patient outcomes. This article reviews the features and outcomes associated with aspiration, stent retrievers, and combination catheters used in current practice. There is also a discussion on clinical considerations based on anatomical features and clot composition. The reperfusion grading scale and outcome metrics commonly used following thrombectomy when a patient is still in the hospital are reviewed. Lastly, there are proposed discharge and outpatient follow-up goals in caring for patients hospitalized for a stroke.

7.
Dis Model Mech ; 14(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-34160002

ABSTRACT

Spinocerebellar ataxia type 7 (SCA7) is an autosomal-dominant neurodegenerative disorder caused by a CAG repeat expansion in the coding region of the ataxin-7 gene. Infantile-onset SCA7 patients display extremely large repeat expansions (>200 CAGs) and exhibit progressive ataxia, dysarthria, dysphagia and retinal degeneration. Severe hypotonia, aspiration pneumonia and respiratory failure often contribute to death in affected infants. To better understand the features of respiratory and upper airway dysfunction in SCA7, we examined breathing and putative phrenic and hypoglossal neuropathology in a knock-in mouse model of early-onset SCA7 carrying an expanded allele with 266 CAG repeats. Whole-body plethysmography was used to measure awake spontaneously breathing SCA7-266Q knock-in mice at baseline in normoxia and during a hypercapnic/hypoxic respiratory challenge at 4 and 8 weeks, before and after the onset of disease. Postmortem studies included quantification of putative phrenic and hypoglossal motor neurons and microglia, and analysis of ataxin-7 aggregation at end stage. SCA7-266Q mice had profound breathing deficits during a respiratory challenge, exhibiting reduced respiratory output and a greater percentage of time in apnea. Histologically, putative phrenic and hypoglossal motor neurons of SCA7 mice exhibited a reduction in number accompanied by increased microglial activation, indicating neurodegeneration and neuroinflammation. Furthermore, intranuclear ataxin-7 accumulation was observed in cells neighboring putative phrenic and hypoglossal motor neurons in SCA7 mice. These findings reveal the importance of phrenic and hypoglossal motor neuron pathology associated with respiratory failure and upper airway dysfunction, which are observed in infantile-onset SCA7 patients and likely contribute to their early death.


Subject(s)
Retinal Degeneration , Spinocerebellar Ataxias , Animals , Ataxin-7 , Disease Models, Animal , Humans , Mice , Nerve Tissue Proteins/genetics , Spinocerebellar Ataxias/complications , Spinocerebellar Ataxias/pathology
9.
Respir Physiol Neurobiol ; 282: 103525, 2020 11.
Article in English | MEDLINE | ID: mdl-32805420

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disorder that results in death due to respiratory failure. Many genetic defects are associated with ALS; one such defect is a mutation in the gene encoding optineurin (OPTN). Using an optineurin null mouse (Optn-/-), we sought to characterize the impact of optineurin deficiency on respiratory neurodegeneration. Respiratory function was assessed at 6 and 12 mo of age using whole body plethysmography at baseline during normoxia (FiO2: 0.21; N2 balance) and during a respiratory challenge with hypoxia and hypercapnia (FiCO2: 0.07, FiO2: 0.10; N2 balance). Histological analyses to assess motor neuron viability and respiratory nerve integrity were performed in the medulla, cervical spinal cord, hypoglossal nerve, and phrenic nerve. Minute ventilation, peak inspiratory flow, and peak expiratory flow are significantly reduced during a respiratory challenge in 6 mo Optn-/-mice. By 12 mo, tidal volume is also significantly reduced in Optn-/- mice. Furthermore, 12mo Optn-/- mice exhibit hypoglossal motor neuron loss, phrenic and hypoglossal dysmyelination, and accumulated mitochondria in the hypoglossal nerve axons. Overall, these data indicate that Optn-/- mice display neurodegenerative respiratory dysfunction and are a useful model to study the impact of novel therapies on respiratory function for optineurin-deficient ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis , Cell Cycle Proteins/deficiency , Hypoglossal Nerve/pathology , Membrane Transport Proteins/deficiency , Mitophagy/physiology , Motor Neurons/pathology , Nerve Degeneration/pathology , Phrenic Nerve/pathology , Respiratory Insufficiency , Amyotrophic Lateral Sclerosis/complications , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Respiratory Insufficiency/etiology , Respiratory Insufficiency/genetics , Respiratory Insufficiency/pathology , Respiratory Insufficiency/physiopathology
10.
Sci Rep ; 10(1): 8967, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32488044

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal neuromuscular disease caused by deleterious mutations in the DMD gene which encodes the dystrophin protein. Skeletal muscle weakness and eventual muscle degradation due to loss of dystrophin are well-documented pathological hallmarks of DMD. In contrast, the neuropathology of this disease remains understudied despite the emerging evidence of neurological abnormalities induced by dystrophin loss. Using quantitative morphological analysis of nerve sections, we characterize axonopathies in the phrenic and hypoglossal (XII) nerves of mdx mice. We observe dysfunction in these nerves - which innervate the diaphragm and genioglossus respectively - that we propose contributes to respiratory failure, the most common cause of death in DMD. These observations highlight the importance in the further characterization of the neuropathology of DMD. Additionally, these observations underscore the necessity in correcting both the nervous system pathology in addition to skeletal muscle deficits to ameliorate this disease.


Subject(s)
Axons/pathology , Dystrophin/genetics , Loss of Function Mutation , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Animals , Diaphragm/innervation , Disease Models, Animal , Hypoglossal Nerve/pathology , Hypoglossal Nerve/physiopathology , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/complications , Phrenic Nerve/pathology , Phrenic Nerve/physiopathology , Respiratory Insufficiency/etiology
11.
Int J Mol Sci ; 21(6)2020 Mar 24.
Article in English | MEDLINE | ID: mdl-32214050

ABSTRACT

Pompe disease is a glycogen storage disease caused by a deficiency in acid α-glucosidase (GAA), a hydrolase necessary for the degradation of lysosomal glycogen. This deficiency in GAA results in muscle and neuronal glycogen accumulation, which causes respiratory insufficiency. Pompe disease mouse models provide a means of assessing respiratory pathology and are important for pre-clinical studies of novel therapies that aim to treat respiratory dysfunction and improve quality of life. This review aims to compile and summarize existing manuscripts that characterize the respiratory phenotype of Pompe mouse models. Manuscripts included in this review were selected utilizing specific search terms and exclusion criteria. Analysis of these findings demonstrate that Pompe disease mouse models have respiratory physiological defects as well as pathologies in the diaphragm, tongue, higher-order respiratory control centers, phrenic and hypoglossal motor nuclei, phrenic and hypoglossal nerves, neuromuscular junctions, and airway smooth muscle. Overall, the culmination of these pathologies contributes to severe respiratory dysfunction, underscoring the importance of characterizing the respiratory phenotype while developing effective therapies for patients.


Subject(s)
Disease Models, Animal , Glycogen Storage Disease Type II/genetics , Phenotype , Respiration , Animals , Glycogen Storage Disease Type II/pathology , Glycogen Storage Disease Type II/physiopathology , Mice
12.
Article in English | MEDLINE | ID: mdl-31893284

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and spinocerebellar ataxia (SCA) are neurodegenerative disorders that result in progressive motor dysfunction and ultimately lead to respiratory failure. Rodent models of neurodegenerative disorders provide a means to study the respiratory motor unit pathology that results in respiratory failure. In addition, they are important for pre-clinical studies of novel therapies that improve breathing, quality of life, and survival. The goal of this review is to compare the respiratory phenotype of two neurodegenerative disorders that have different pathological origins, but similar physiological outcomes. Manuscripts reviewed were identified using specific search terms and exclusion criteria. We excluded manuscripts that investigated novel therapeutics and only included those manuscripts that describe the respiratory pathology. The ALS manuscripts describe pathology in respiratory physiology, the phrenic and hypoglossal motor units, respiratory neural control centers, and accessory respiratory muscles. The SCA rodent model manuscripts characterized pathology in overall respiratory function, phrenic motor units and hypoglossal motor neurons. Overall, a combination of pathology in the respiratory motor units and control centers contribute to devastating respiratory dysfunction.

13.
Clin Immunol ; 175: 16-25, 2017 02.
Article in English | MEDLINE | ID: mdl-27915003

ABSTRACT

Ataxia-Teleangiectasia (A-T) is a neurodegenerative disorder due to mutations in ATM gene. ATM in the nucleus ensures DNA repair, while its role in the cytosol is still poorly clarified. Abnormal autophagy has been documented in other neurodegenerative disorders, thus we evaluated whether alteration in this process may be involved in the pathogenesis of A-T by analyzing the autophagic vesicles and the genes implicated in the different stages of autophagy. Through transmission electron microscopy (TEM) and immunofluorescence analysis we observed an accumulation of APs associated with a LC3 puncta pattern, and a reduced number of ALs. We also documented an increased expression of genes involved in AP and lysosome biogenesis and function, and a decrease of Vps18 expression, involved in their vesicular trafficking and fusion. mTORC1-controlled proteins were hyperphosphorylated in A-T, in keeping with an increased mTOR inhibitory influence of autophagy. Betamethasone is able to promote the degradation of SQSTM1, a biomarker of autophagy. Collectively, our results indicate that in cells from A-T patients, the APs maturation is active, while the fusion between APs and lysosomes is inappropriate, thus implying abnormalities in the cell-clearance process. We also documented a positive effect of Betamethasone on molecules implicated in autophagosome degradation.


Subject(s)
Ataxia/pathology , Autophagy/physiology , Lymphocytes/pathology , Neurodegenerative Diseases/pathology , Ataxia/drug therapy , Ataxia/metabolism , Autophagy/drug effects , Betamethasone/therapeutic use , Biomarkers/metabolism , Cells, Cultured , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Lymphocytes/drug effects , Lymphocytes/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Sequestosome-1 Protein/metabolism , TOR Serine-Threonine Kinases/metabolism
14.
Int Rev Immunol ; 35(1): 25-38, 2016.
Article in English | MEDLINE | ID: mdl-25774666

ABSTRACT

Primary immunodeficiencies (PIDs) include a heterogeneous group of mostly monogenic diseases characterized by functional/developmental alterations of the immune system. Skin and skin annexa abnormalities may be a warning sign of immunodeficiency, since both epidermal and thymic epithelium have ectodermal origin. In this review, we will focus on the most common immune disorders associated with ectodermal alterations. Elevated IgE levels represent the immunological hallmark of hyper-IgE syndrome, characterized by severe eczema and susceptibility to infections. Ectodermal dysplasia (ED) is a group of rare disorders that affect tissues of ectodermal origin. Hypoidrotic ED (HED), the most common form, is inherited as autosomal dominant, autosomal recessive or X-linked trait (XLHED). HED and XLHED are caused by mutations in NEMO and EDA-1 genes, respectively, and show similarities in the cutaneous involvement but differences in the susceptibility to infections and immunological phenotype. Alterations in the transcription factor FOXN1 gene, expressed in the mature thymic and skin epithelia, are responsible for human and murine athymia and prevent the development of the T-cell compartment associated to ectodermal abnormalities such as alopecia and nail dystrophy. The association between developmental abnormalities of the skin and immunodeficiencies suggest a role of the skin as a primary lymphoid organ. Recently, it has been demonstrated that a co-culture of human skin-derived keratinocytes and fibroblasts, in the absence of thymic components, can support the survival of human haematopoietic stem cells and their differentiation into T-lineage committed cells.


Subject(s)
DiGeorge Syndrome/immunology , Ectodermal Dysplasia/immunology , Job Syndrome/immunology , Severe Combined Immunodeficiency/immunology , Skin/immunology , T-Lymphocytes/immunology , Animals , DiGeorge Syndrome/genetics , Ectodermal Dysplasia/genetics , Eczema/immunology , Epidermis/growth & development , Epidermis/immunology , Humans , Job Syndrome/genetics , Mice , Mutation , Phenotype , Severe Combined Immunodeficiency/genetics , T-Lymphocytes/metabolism , Thymus Gland/growth & development , Thymus Gland/immunology
15.
Int Rev Immunol ; 33(2): 83-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24432845

ABSTRACT

FOXN1 gene belongs to the forkhead box gene family that comprises a diverse group of "winged-helix" transcription factors that have been implicated in a variety of biochemical and cellular processes, such as development, metabolism, aging and cancer. These transcription factors share the common property of being developmentally regulated and of directing tissue-specific transcription and cell-fate decisions. Foxn1 is selectively expressed in thymic and skin epithelial cells, where it acts through its molecular targets to regulate the balance between growth and differentiation. In particular, Foxn1 is required for thymic epithelial patterning and differentiation from the initial epithelial thymic anlage to a functional cortical and medullary thymic epithelial cells (TECs) meshwork necessary for the crosstalk with the lymphoid compartment. A mutation in FoxN1 generates alymphoid cystic thymic dysgenesis due to defective TECs, causing primary T-cell immunodeficiency, named Nude/SCID syndrome, and leads to a hairless "nude" phenotype in both mice and humans. This immune defect represents the first example of a Severe Combined Immunodeficiencies (SCID) phenotype not primarily related to an abnormality intrinsic of the hematopoietic cell, but rather to a peculiar alteration of the thymic epithelia cell. This review focuses on the key role of FOXN1 in cell development and its clinical implication in humans.


Subject(s)
Epithelial Cells/physiology , Forkhead Transcription Factors/physiology , Immunologic Deficiency Syndromes/genetics , Thymus Gland/immunology , Animals , Cell Differentiation , Cell Growth Processes , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Nude , Mice, SCID , Mutation/genetics , Organ Specificity , Organogenesis
16.
PLoS One ; 8(12): e81786, 2013.
Article in English | MEDLINE | ID: mdl-24349129

ABSTRACT

The thymus is the primary organ able to support T cell ontogeny, abrogated in FOXN1(-/-) human athymia. Although evidence indicates that in animal models T lymphocytes may differentiate at extrathymic sites, whether this process is really thymus-independent has still to be clarified. In an athymic FOXN1(-/-) fetus, in which we previously described a total blockage of CD4(+) and partial blockage of CD8(+) cell development, we investigated whether intestine could play a role as extrathymic site of T-lymphopoiesis in humans. We document the presence of few extrathymically developed T lymphocytes and the presence in the intestine of CD3(+) and CD8(+), but not of CD4(+) cells, a few of them exhibiting a CD45RA(+) naïve phenotype. The expression of CD3εεpTα, RAG1 and RAG2 transcripts in the intestine and TCR gene rearrangement was also documented, thus indicating that in humans the partial T cell ontogeny occurring at extrathymic sites is a thymus- and FOXN1-independent process.


Subject(s)
Forkhead Transcription Factors/deficiency , Intestines/cytology , T-Lymphocyte Subsets/pathology , Thymus Gland/pathology , Antigens, CD/genetics , Antigens, CD/immunology , CD3 Complex/genetics , CD3 Complex/immunology , CD4 Antigens/genetics , CD4 Antigens/immunology , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Fetus , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression , Gene Silencing , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Immunophenotyping , Intestines/immunology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Nuclear Proteins/genetics , Nuclear Proteins/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/abnormalities , Thymus Gland/immunology
17.
Int Immunol ; 25(12): 703-14, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24038600

ABSTRACT

In humans, the thymus is the primary lymphoid organ able to support the development of T cells through its three-dimensional (3D) organization of the thymic stromal cells. Since a remarkable number of similarities are shared between the thymic epithelial cells (TECs) and skin-derived keratinocytes and fibroblasts, in this study we used human keratinocytes seeded with fibroblasts on the 3D poly ε-caprolactone scaffold to evaluate their ability to replace TECs in supporting T-cell differentiation from human haematopoietic stem cells (HSCs). We observed that in the multicellular biocomposite, early thymocytes expressing CD7(+)CD1a(+), peculiar markers of an initial T-cell commitment, were de novo generated. Molecular studies of genes selectively expressed during T-cell development revealed that TAL1 was down-regulated and Spi-B was up-regulated in the cell suspension, consistently with a T-cell lineage commitment. Moreover, PTCRA and RAG2 expression was detected, indicative of a recombinant activity, required for the generation of a T-cell receptor repertoire. Our results indicate that in the multicellular biocomposite, containing skin-derived elements in the absence of thymic stroma, HSCs do start differentiating toward a T-cell lineage commitment. In conclusion, the construct described in this study exerts some properties of a lymphoid organoid, suitable for future clinical applications in cell-based therapies.


Subject(s)
Caproates , Cell Differentiation , Fibroblasts/physiology , Hematopoietic Stem Cells/cytology , Keratinocytes/physiology , Lactones , Precursor Cells, T-Lymphoid/cytology , Tissue Scaffolds , Caproates/chemistry , Cell Communication , Cell Proliferation , Cell Survival , Coculture Techniques , Epidermal Cells , Fibroblasts/ultrastructure , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Keratinocytes/ultrastructure , Lactones/chemistry , Leukocytes, Mononuclear/physiology , Phenotype , Porosity , Precursor Cells, T-Lymphoid/metabolism , Thymus Gland/cytology , Thymus Gland/physiology , Tissue Scaffolds/chemistry
18.
Arterioscler Thromb Vasc Biol ; 33(10): 2415-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23950144

ABSTRACT

OBJECTIVE: The role of endothelial G protein-coupled receptor kinase 2 (GRK2) was investigated in mice with selective deletion of the kinase in the endothelium (Tie2-CRE/GRK2(fl/fl)). APPROACH AND RESULTS: Aortas from Tie2-CRE/GRK2(fl/fl) presented functional and structural alterations as compared with control GRK2(fl/fl) mice. In particular, vasoconstriction was blunted to different agonists, and collagen and elastic rearrangement and macrophage infiltration were observed. In primary cultured endothelial cells deficient for GRK2, mitochondrial reactive oxygen species was increased, leading to expression of cytokines. Chronic treatment with a reactive oxygen species scavenger in mice corrected the vascular phenotype by recovering vasoconstriction, structural abnormalities, and reducing macrophage infiltration. CONCLUSIONS: These results demonstrate that GRK2 removal compromises vascular phenotype and integrity by increasing endothelial reactive oxygen species production.


Subject(s)
Aorta, Thoracic/enzymology , Endothelial Cells/enzymology , G-Protein-Coupled Receptor Kinase 2/metabolism , Reactive Oxygen Species/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Cells, Cultured , Collagen/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Elastic Tissue/metabolism , Elastic Tissue/pathology , Endothelial Cells/pathology , Free Radical Scavengers/pharmacology , G-Protein-Coupled Receptor Kinase 2/deficiency , G-Protein-Coupled Receptor Kinase 2/genetics , Homeostasis , Inflammation Mediators/metabolism , Integrases/genetics , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Phenotype , Receptor, TIE-2/genetics , Signal Transduction , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
19.
Front Immunol ; 4: 187, 2013.
Article in English | MEDLINE | ID: mdl-23874334

ABSTRACT

T cell ontogeny is a sophisticated process, which takes place within the thymus through a series of well-defined discrete stages. The process requires a proper lympho-stromal interaction. In particular, cortical and medullary thymic epithelial cells (cTECs, mTECs) drive T cell differentiation, education, and selection processes, while the thymocyte-dependent signals allow thymic epithelial cells (TECs) to maturate and provide an appropriate thymic microenvironment. Alterations in genes implicated in thymus organogenesis, including Tbx1, Pax1, Pax3, Pax9, Hoxa3, Eya1, and Six1, affect this well-orchestrated process, leading to disruption of thymic architecture. Of note, in both human and mice, the primordial TECs are yet unable to fully support T cell development and only after the transcriptional activation of the Forkhead-box n1 (FOXN1) gene in the thymic epithelium this essential function is acquired. FOXN1 is a master regulator in the TEC lineage specification in that it down-stream promotes transcription of genes, which, in turn, regulate TECs differentiation. In particular, FOXN1 mainly regulates TEC patterning in the fetal stage and TEC homeostasis in the post-natal thymus. An inborn null mutation in FOXN1 leads to Nude/severe combined immunodeficiency (SCID) phenotype in mouse, rat, and humans. In Foxn1 (-/-) nude animals, initial formation of the primordial organ is arrested and the primordium is not colonized by hematopoietic precursors, causing a severe primary T cell immunodeficiency. In humans, the Nude/SCID phenotype is characterized by congenital alopecia of the scalp, eyebrows, and eyelashes, nail dystrophy, and a severe T cell immunodeficiency, inherited as an autosomal recessive disorder. Aim of this review is to summarize all the scientific information so far available to better characterize the pivotal role of the master regulator FOXN1 transcription factor in the TEC lineage specifications and functionality.

20.
Ital J Pediatr ; 39: 12, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23410226

ABSTRACT

Palamaro et al. describe a child with recurrent bronchopneumonia and very high IgE levels in which a variation, R156H, was found in the IL12RB1 gene that encodes the IL-12Rß1 chain. Based on the absence of this variation in 50 unrelated individuals they conclude it is a mutation. We (van de Vosse and van Dissel) feel there is no reason to suspect a defect in IL-12 signaling based on the clinical data, nor evidence for a functional defect in IL-12 signaling in this patient. In addition, the variation is not novel and known as a polymorphism. Without any functional evidence that R156H is a mutation, the current claim is not substantiated. Palamaro et al. respond to argue that the amino acid substitution, R156H described in the described case exerts a summatory effect, as a genetic cofactor, along with an additional and still unidentified molecular alteration of the same pathway.


Subject(s)
Bronchopneumonia/immunology , Immunoglobulin E/blood , Receptors, Interleukin-12/deficiency , Female , Humans
SELECTION OF CITATIONS
SEARCH DETAIL