Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Elife ; 102021 06 04.
Article En | MEDLINE | ID: mdl-34085925

Defective autophagy is strongly associated with chronic inflammation. Loss-of-function of the core autophagy gene Atg16l1 increases risk for Crohn's disease in part by enhancing innate immunity through myeloid cells such as macrophages. However, autophagy is also recognized as a mechanism for clearance of certain intracellular pathogens. These divergent observations prompted a re-evaluation of ATG16L1 in innate antimicrobial immunity. In this study, we found that loss of Atg16l1 in myeloid cells enhanced the killing of virulent Shigella flexneri (S.flexneri), a clinically relevant enteric bacterium that resides within the cytosol by escaping from membrane-bound compartments. Quantitative multiplexed proteomics of murine bone marrow-derived macrophages revealed that ATG16L1 deficiency significantly upregulated proteins involved in the glutathione-mediated antioxidant response to compensate for elevated oxidative stress, which simultaneously promoted S.flexneri killing. Consistent with this, myeloid-specific deletion of Atg16l1 in mice accelerated bacterial clearance in vitro and in vivo. Pharmacological induction of oxidative stress through suppression of cysteine import enhanced microbial clearance by macrophages. Conversely, antioxidant treatment of macrophages permitted S.flexneri proliferation. These findings demonstrate that control of oxidative stress by ATG16L1 and autophagy regulates antimicrobial immunity against intracellular pathogens.


Autophagy-Related Proteins/deficiency , Autophagy , Dysentery, Bacillary/microbiology , Immunity, Innate , Macrophages/microbiology , Oxidative Stress , Proteome , Proteomics , Shigella flexneri/pathogenicity , Animals , Autophagy-Related Proteins/genetics , Cells, Cultured , Disease Models, Animal , Dysentery, Bacillary/immunology , Dysentery, Bacillary/metabolism , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microbial Viability , Shigella flexneri/immunology , Shigella flexneri/metabolism , Virulence
2.
Nature ; 587(7833): 275-280, 2020 11.
Article En | MEDLINE | ID: mdl-32971525

Mutations in the death receptor FAS1,2 or its ligand FASL3 cause autoimmune lymphoproliferative syndrome, whereas mutations in caspase-8 or its adaptor FADD-which mediate cell death downstream of FAS and FASL-cause severe immunodeficiency in addition to autoimmune lymphoproliferative syndrome4-6. Mouse models have corroborated a role for FADD-caspase-8 in promoting inflammatory responses7-12, but the mechanisms that underlie immunodeficiency remain undefined. Here we identify NEDD4-binding protein 1 (N4BP1) as a suppressor of cytokine production that is cleaved and inactivated by caspase-8. N4BP1 deletion in mice increased the production of select cytokines upon stimulation of the Toll-like receptor (TLR)1-TLR2 heterodimer (referred to herein as TLR1/2), TLR7 or TLR9, but not upon engagement of TLR3 or TLR4. N4BP1 did not suppress TLR3 or TLR4 responses in wild-type macrophages, owing to TRIF- and caspase-8-dependent cleavage of N4BP1. Notably, the impaired production of cytokines in response to TLR3 and TLR4 stimulation of caspase-8-deficient macrophages13 was largely rescued by co-deletion of N4BP1. Thus, the persistence of intact N4BP1 in caspase-8-deficient macrophages impairs their ability to mount robust cytokine responses. Tumour necrosis factor (TNF), like TLR3 or TLR4 agonists, also induced caspase-8-dependent cleavage of N4BP1, thereby licensing TRIF-independent TLRs to produce higher levels of inflammatory cytokines. Collectively, our results identify N4BP1 as a potent suppressor of cytokine responses; reveal N4BP1 cleavage by caspase-8 as a point of signal integration during inflammation; and offer an explanation for immunodeficiency caused by mutations of FADD and caspase-8.


Caspase 8/metabolism , Cytokines/immunology , Immunity, Innate/immunology , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cells, Cultured , Cytokines/antagonists & inhibitors , Humans , Inflammation/immunology , Mice , Mice, Inbred C57BL , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
3.
Eur J Immunol ; 50(6): 891-902, 2020 06.
Article En | MEDLINE | ID: mdl-32043568

CD96 is a member of the poliovirus receptor (PVR, CD155)-nectin family that includes T cell Ig and ITIM domain (TIGIT) and CD226. While CD96, TIGIT, and CD226 have important roles in regulating NK cell activity, and TIGIT and CD226 have also been shown to regulate T cell responses, it is unclear whether CD96 has inhibitory or stimulatory function in CD8+ T cells. Here, we demonstrate that CD96 has co-stimulatory function on CD8+ T cells. Crosslinking of CD96 on human or mouse CD8+ T cells induced activation, effector cytokine production, and proliferation. CD96 was found to transduce its activating signal through the MEK-ERK pathway. CD96-mediated signaling led to increased frequencies of NUR77- and T-bet-expressing CD8+ T cells and enhanced cytotoxic effector activity, indicating that CD96 can modulate effector T cell differentiation. Antibody blockade of CD96 or genetic ablation of CD96 expression on CD8+ T cells impaired expression of transcription factors and proinflammatory cytokines associated with CD8+ T cell activation in in vivo models. Taken together, CD96 has a co-stimulatory role in CD8+ T cell activation and effector function.


Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Lymphocyte Activation , MAP Kinase Signaling System/immunology , Models, Immunological , Animals , Antigens, CD/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Humans , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout
4.
J Leukoc Biol ; 107(6): 941-952, 2020 06.
Article En | MEDLINE | ID: mdl-31985117

Receptor interacting protein kinase 1 (RIP1) is a critical effector of inflammatory responses and cell death activation. Cell death pathways regulated by RIP1 include caspase-dependent apoptosis and caspase-independent necroptosis. The kinase activity of RIP1 has been associated with a number of inflammatory, neurodegenerative, and oncogenic diseases. In this study, we use the RIP1 kinase inhibitor GNE684 to demonstrate that RIP1 inhibition can effectively block skin inflammation and immune cell infiltrates in livers of Sharpin mutant (Cpdm; chronic proliferative dermatitis) mice in an interventional setting, after disease onset. On the other hand, genetic inactivation of RIP1 (RIP1 KD) or ablation of RIP3 (RIP3 KO) or MLKL (MLKL KO) did not affect testicular pathology of aging male mice. Likewise, infection with vaccinia virus or with mouse gammaherpesvirus MHV68 resulted in similar viral clearance in wild-type, RIP1 KD, and RIP3 KO mice. In summary, this study highlights the benefits of inhibiting RIP1 in skin inflammation, as opposed to its lack of relevance for testicular longevity and the response to certain viral infections.


Dermatitis/genetics , Herpesviridae Infections/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Skin/immunology , Vaccinia/genetics , Animals , Chronic Disease , Dermatitis/immunology , Dermatitis/pathology , Dermatitis/virology , Disease Models, Animal , Gammaherpesvirinae/immunology , Gammaherpesvirinae/pathogenicity , Gene Expression Regulation , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Inflammation , Liver/immunology , Liver/pathology , Liver/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Protein Kinases/deficiency , Protein Kinases/genetics , Protein Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Signal Transduction , Skin/pathology , Skin/virology , Testis/immunology , Testis/pathology , Testis/virology , Vaccinia/immunology , Vaccinia/pathology , Vaccinia/virology , Vaccinia virus/immunology , Vaccinia virus/pathogenicity , Virus Replication/immunology
5.
Cell Death Differ ; 27(1): 161-175, 2020 01.
Article En | MEDLINE | ID: mdl-31101885

The kinase RIP1 acts in multiple signaling pathways to regulate inflammatory responses and it can trigger both apoptosis and necroptosis. Its kinase activity has been implicated in a range of inflammatory, neurodegenerative, and oncogenic diseases. Here, we explore the effect of inhibiting RIP1 genetically, using knock-in mice that express catalytically inactive RIP1 D138N, or pharmacologically, using the murine-potent inhibitor GNE684. Inhibition of RIP1 reduced collagen antibody-induced arthritis, and prevented skin inflammation caused by mutation of Sharpin, or colitis caused by deletion of Nemo from intestinal epithelial cells. Conversely, inhibition of RIP1 had no effect on tumor growth or survival in pancreatic tumor models driven by mutant Kras, nor did it reduce lung metastases in a B16 melanoma model. Collectively, our data emphasize a role for the kinase activity of RIP1 in certain inflammatory disease models, but question its relevance to tumor progression and metastases.


Inflammation/enzymology , Neoplasms/enzymology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Arthritis/enzymology , Cell Death , Cell Line , Cell Line, Tumor , Colitis/etiology , Colitis/prevention & control , Dermatitis/enzymology , Female , Gene Knock-In Techniques , Humans , Ileitis/etiology , Ileitis/prevention & control , Intracellular Signaling Peptides and Proteins/genetics , Male , Melanoma, Experimental/pathology , Mice , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology
6.
Cell Rep ; 27(9): 2679-2689.e3, 2019 05 28.
Article En | MEDLINE | ID: mdl-31141691

Cellular inhibitor of apoptosis proteins cIAP1 and cIAP2 ubiquitinate nuclear factor κB (NF-κB)-inducing kinase (NIK) to suppress non-canonical NF-κB signaling and substrates such as receptor interacting protein kinase 1 (RIPK1) to promote cell survival. We investigate how these functions contribute to homeostasis by eliminating cIap2 from adult cIap1-deficient mice. cIAP1 and cIAP2 (cIAP1/2) deficiency causes rapid weight loss and inflammation, with aberrant cell death, indicated by cleaved caspases-3 and -8, prevalent in intestine and liver. Deletion of Casp8 and Ripk3 prevents this aberrant cell death, reduces the inflammation, and prolongs mouse survival, whereas Ripk3 loss alone offers little benefit. Residual inflammation in mice lacking cIap1/2, Casp8, and Ripk3 is reduced by inhibition of NIK. Loss of Casp8 and Mlkl (mixed lineage kinase domain-like), but not Mlkl loss alone, also prevents cIAP1/2-deficient mice from dying around embryonic day 11. Therefore, a major function of cIAP1/2 in vivo is to suppress caspase-8-dependent cell death.


Apoptosis , Baculoviral IAP Repeat-Containing 3 Protein/physiology , Caspase 8/metabolism , Inflammation/prevention & control , Inhibitor of Apoptosis Proteins/physiology , NF-kappa B/metabolism , Animals , Caspase 8/genetics , Female , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , Signal Transduction , Ubiquitin/metabolism
7.
Sci Transl Med ; 10(468)2018 11 21.
Article En | MEDLINE | ID: mdl-30463918

Preclinical and clinical evidence indicates that a subset of asthma is driven by type 2 cytokines such as interleukin-4 (IL-4), IL-5, IL-9, and IL-13. Additional evidence predicts pathogenic roles for IL-6 and type I and type II interferons. Because each of these cytokines depends on Janus kinase 1 (JAK1) for signal transduction, and because many of the asthma-related effects of these cytokines manifest in the lung, we hypothesized that lung-restricted JAK1 inhibition may confer therapeutic benefit. To test this idea, we synthesized iJak-381, an inhalable small molecule specifically designed for local JAK1 inhibition in the lung. In pharmacodynamic models, iJak-381 suppressed signal transducer and activator of transcription 6 activation by IL-13. Furthermore, iJak-381 suppressed ovalbumin-induced lung inflammation in both murine and guinea pig asthma models and improved allergen-induced airway hyperresponsiveness in mice. In a model driven by human allergens, iJak-381 had a more potent suppressive effect on neutrophil-driven inflammation compared to systemic corticosteroid administration. The inhibitor iJak-381 reduced lung pathology, without affecting systemic Jak1 activity in rodents. Our data show that local inhibition of Jak1 in the lung can suppress lung inflammation without systemic Jak inhibition in rodents, suggesting that this strategy might be effective for treating asthma.


Asthma/drug therapy , Asthma/enzymology , Janus Kinase 1/antagonists & inhibitors , Lung/enzymology , Protein Kinase Inhibitors/therapeutic use , Administration, Inhalation , Allergens , Animals , Asthma/pathology , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/metabolism , Eosinophils/pathology , Guinea Pigs , Inflammation/pathology , Janus Kinase 1/metabolism , Lung/drug effects , Lung/pathology , Ovalbumin , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Signal Transduction , Treatment Outcome
8.
Nat Immunol ; 19(3): 246-254, 2018 03.
Article En | MEDLINE | ID: mdl-29358708

Defective autophagy is linked to diseases such as rheumatoid arthritis, lupus and inflammatory bowel disease (IBD). However, the mechanisms by which autophagy limits inflammation remain poorly understood. Here we found that loss of the autophagy-related gene Atg16l1 promoted accumulation of the adaptor TRIF and downstream signaling in macrophages. Multiplex proteomic profiling identified SQSTM1 and Tax1BP1 as selective autophagy-related receptors that mediated the turnover of TRIF. Knockdown of Tax1bp1 increased production of the cytokines IFN-ß and IL-1ß. Mice lacking Atg16l1 in myeloid cells succumbed to lipopolysaccharide-mediated sepsis but enhanced their clearance of intestinal Salmonella typhimurium in an interferon receptor-dependent manner. Human macrophages with the Crohn's disease-associated Atg16l1 variant T300A exhibited more production of IFN-ß and IL-1ß. An elevated interferon-response gene signature was observed in patients with IBD who were resistant to treatment with an antibody to the cytokine TNF. These findings identify selective autophagy as a key regulator of signaling via the innate immune system.


Adaptor Proteins, Vesicular Transport/immunology , Autophagy/immunology , Immunity, Innate/immunology , Inflammation/immunology , Animals , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/immunology , Crohn Disease/immunology , Female , Humans , Macrophages/immunology , Male , Mice , Mice, Transgenic , Signal Transduction/immunology
9.
Nat Commun ; 9(1): 179, 2018 01 12.
Article En | MEDLINE | ID: mdl-29330524

NF-κB-inducing kinase (NIK) mediates non-canonical NF-κB signaling downstream of multiple TNF family members, including BAFF, TWEAK, CD40, and OX40, which are implicated in the pathogenesis of systemic lupus erythematosus (SLE). Here, we show that experimental lupus in NZB/W F1 mice can be treated with a highly selective and potent NIK small molecule inhibitor. Both in vitro as well as in vivo, NIK inhibition recapitulates the pharmacological effects of BAFF blockade, which is clinically efficacious in SLE. Furthermore, NIK inhibition also affects T cell parameters in the spleen and proinflammatory gene expression in the kidney, which may be attributable to inhibition of OX40 and TWEAK signaling, respectively. As a consequence, NIK inhibition results in improved survival, reduced renal pathology, and lower proteinuria scores. Collectively, our data suggest that NIK inhibition is a potential therapeutic approach for SLE.


B-Lymphocytes/drug effects , Kidney/drug effects , Lupus Erythematosus, Systemic/immunology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , T-Lymphocytes/drug effects , Animals , B-Lymphocytes/immunology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokine TWEAK/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Gene Expression/drug effects , Humans , In Vitro Techniques , Inflammation/genetics , Interleukin-12 Subunit p40/drug effects , Interleukin-12 Subunit p40/immunology , Kidney/immunology , Kidney/pathology , Lupus Erythematosus, Systemic/drug therapy , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Mice , Mice, Inbred NZB , Molecular Targeted Therapy , Proteinuria/immunology , Receptors, OX40/metabolism , Signal Transduction , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/immunology , NF-kappaB-Inducing Kinase
10.
Immunohorizons ; 2(5): 164-171, 2018 05 30.
Article En | MEDLINE | ID: mdl-31022698

Intestinal epithelial cells form a physical barrier that is tightly regulated to control intestinal permeability. Proinflammatory cytokines, such as TNF-α, increase epithelial permeability through disruption of epithelial junctions. The regulation of the epithelial barrier in inflammatory gastrointestinal disease remains to be fully characterized. In this article, we show that the human inflammatory bowel disease genetic susceptibility gene C1ORF106 plays a key role in regulating gut epithelial permeability. C1ORF106 directly interacts with cytohesins to maintain functional epithelial cell junctions. C1orf106-deficient mice are hypersensitive to TNF-α-induced increase in epithelial permeability, and this is associated with increased diarrhea. This study identifies C1ORF106 as an epithelial cell junction protein, and the loss of C1ORF106 augments TNF-α-induced intestinal epithelial leakage and diarrhea that may play a critical role in the development of inflammatory bowel disease.


Carrier Proteins/genetics , Inflammatory Bowel Diseases/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Animals , Caco-2 Cells , Carrier Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Epithelial Cells/metabolism , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Permeability , Receptors, Cytoplasmic and Nuclear/metabolism , Tight Junctions/genetics , Tight Junctions/metabolism , Tumor Necrosis Factor-alpha/genetics
11.
Sci Signal ; 10(475)2017 Apr 18.
Article En | MEDLINE | ID: mdl-28420753

Tumor progression locus 2 (TPL2; also known as MAP3K8) is a mitogen-activated protein kinase (MAPK) kinase kinase (MAP3K) that phosphorylates the MAPK kinases MEK1 and MEK2 (MEK1/2), which, in turn, activate the MAPKs extracellular signal-regulated kinase 1 (ERK1) and ERK2 (ERK1/2) in macrophages stimulated through the interleukin-1 receptor (IL-1R), Toll-like receptors (TLRs), or the tumor necrosis factor receptor (TNFR). We describe a conserved and critical role for TPL2 in mediating the effector functions of neutrophils through the activation of the p38 MAPK signaling pathway. Gene expression profiling and functional studies of neutrophils and monocytes revealed a MEK1/2-independent branch point downstream of TPL2 in neutrophils. Biochemical analyses identified the MAPK kinases MEK3 and MEK6 and the MAPKs p38α and p38δ as downstream effectors of TPL2 in these cells. Genetic ablation of the catalytic activity of TPL2 or therapeutic intervention with a TPL2-specific inhibitor reduced the production of inflammatory mediators by neutrophils in response to stimulation with the TLR4 agonist lipopolysaccharide (LPS) in vitro, as well as in rodent models of inflammatory disease. Together, these data suggest that TPL2 is a drug target that activates not only MEK1/2-dependent but also MEK3/6-dependent signaling to promote inflammatory responses.


MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 3/metabolism , Neutrophil Activation , Neutrophils/enzymology , Proto-Oncogene Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Enzyme Activation , Inflammation/enzymology , Inflammation/genetics , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , MAP Kinase Kinase Kinases/genetics , Mice , Mitogen-Activated Protein Kinase 3/genetics , Proto-Oncogene Proteins/genetics , p38 Mitogen-Activated Protein Kinases/genetics
12.
Neurosci Lett ; 506(2): 208-13, 2012 Jan 11.
Article En | MEDLINE | ID: mdl-22100909

Multiple studies converge to implicate alterations of the hippocampus and amygdala in the pathology of autism. We have previously reported anatomical alterations of the meninges, vasculature and fractones, the specialized extracellular matrix (ECM) of the subventricular zone, in the forebrain of adult BTBR T+ tf/J mice, animal model for autism. Here, we used bisbenzidine cell nucleus staining and dual immunofluorescence histochemistry for laminin and N-sulfated heparan sulfate proteoglycans (NS-HSPG) to examine a series of brain sections containing the amygdala and hippocampus in the adult BTBR T+ tf/j mouse. We observed an excessive separation of the two hippocampi, a modified trajectory of the meninges leading to a shrunken choroid plexus in the lateral ventricle, a shorter granular layer of the dentate gyrus, and a reduced size of the amygdala nuclei. The lateral ventricle near the amygdala, and the third ventricle were shrunken. The number and size of fractones, and their immunoreactivity for NS-HSPG, were reduced throughout the third and lateral ventricles walls. Enlarged blood vessels were found at the endopiriform cortex/amygdala interface. These results show anatomical alterations of the hippocampal/amygdala that are associated with defects of the choroid plexus/ventricular system and the ECM in the BTBR T+ TF/J mouse. Similar alterations of the hippocampus/amygdala axis in humans with autism to these observed in BTBR T+ tf/J mice make this animal model highly valuable for the study of autism. Moreover, the meningo/vascular and ECM alterations in BTBR T+ Tf/J mice suggest a possible role of the brain connective tissue in autism.


Amygdala/pathology , Autistic Disorder/pathology , Extracellular Matrix/pathology , Hippocampus/pathology , Amygdala/metabolism , Animals , Autistic Disorder/metabolism , Cerebral Ventricles/metabolism , Cerebral Ventricles/pathology , Disease Models, Animal , Female , Fluorescent Antibody Technique , Heparitin Sulfate/analysis , Hippocampus/metabolism , Male , Mice , Mice, Neurologic Mutants
13.
Neurosci Lett ; 498(3): 173-8, 2011 Jul 12.
Article En | MEDLINE | ID: mdl-21600960

Autism spectrum disorders are characterized by impaired social and communication skills and seem to result from altered neural development. We sought to determine whether the anatomy of the meninges and extracellular matrix (ECM) is altered in an animal model of autism, the BTBR T+ tf/J mouse. This mouse displays white matter tract anatomical defects and exhibits several symptoms of autism. Immunofluorescence cytochemistry for laminin, a major ECM marker, was performed on series of coronal sections of the adult BTBR T+ tf/J brain and the anatomy was analyzed in comparison to B6 wild type mice. Laminin immunoreactivity visualized meninges, blood vessels and the subventricular zone (SVZ) stem cell-associated ECM structures, which I have named fractones. All BTBR T+ tf/J mice observed showed the same forebrain defects. The lateral ventricle volume was severely reduced, the falx cerebri elongated, the arteries enlarged and the choroid plexus atrophied. Compared to B6 mice, fractone numbers in BTBR T+ tf/J mice were reduced by a factor three in the SVZ of the anterior portion of the lateral ventricle. This represents the primary neurogenic zone during adulthood. Fractones were reduced by a factor 1.5 in posterior portions of the lateral ventricle. Moreover, fractone size was reduced throughout the lateral ventricle SVZ. These results show hitherto unsuspected alterations in connective tissue/vasculature and associated ECM in the adult BTBR T+ tf/J mouse. The drastic changes of the connective tissue and ECM in the neurogenic zone of the lateral ventricle may contribute to incorrect neurogenesis during developmental and adult stages.


Autistic Disorder/pathology , Autistic Disorder/physiopathology , Brain/pathology , Choroid Plexus/pathology , Disease Models, Animal , Extracellular Matrix/pathology , Meninges/pathology , Neurogenesis/physiology , Animals , Atrophy/pathology , Extracellular Matrix/metabolism , Female , Laminin/metabolism , Lateral Ventricles/pathology , Male , Mice , Mice, Inbred Strains , Mice, Neurologic Mutants , Prosencephalon/blood supply , Prosencephalon/pathology
...