Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 443
1.
J Cell Physiol ; 238(3): 631-646, 2023 03.
Article En | MEDLINE | ID: mdl-36727620

A common adverse response to the clinical use of glucocorticoids (GCs) is elevated intraocular pressure (IOP) which is a major risk factor for glaucoma. Elevated IOP arises due to impaired outflow of aqueous humor (AH) through the trabecular meshwork (TM). Although GC-induced changes in actin cytoskeletal dynamics, contractile characteristics, and cell adhesive interactions of TM cells are believed to influence AH outflow and IOP, the molecular mechanisms mediating changes in these cellular characteristics are poorly understood. Our studies focused on evaluating changes in the cytoskeletal and cytoskeletal-associated protein (cytoskeletome) profile of human TM cells treated with dexamethasone (Dex) using label-free mass spectrometric quantification, identified elevated levels of specific proteins known to regulate actin stress fiber formation, contraction, actin networks crosslinking, cell adhesion, and Wnt signaling, including LIMCH1, ArgBP2, CNN3, ITGBL1, CTGF, palladin, FAT1, DIAPH2, EPHA4, SIPA1L1, and GPC4. Several of these proteins colocalized with the actin cytoskeleton and underwent alterations in distribution profile in TM cells treated with Dex, and an inhibitor of Abl/Src kinases. Wnt/Planar Cell Polarity (PCP) signaling agonists-Wnt5a and 5b were detected prominently in the cytoskeletome fraction of TM cells, and studies using siRNA to suppress expression of glypican-4 (GPC4), a known modulator of the Wnt/PCP pathway revealed that GPC4 deficiency impairs Dex induced actin stress fiber formation, and activation of c-Jun N-terminal Kinase (JNK) and Rho kinase. Additionally, while Dex augmented, GPC4 deficiency suppressed the formation of actin stress fibers in TM cells in the presence of Dex and Wnt5a. Taken together, these results identify the GPC4-dependent Wnt/PCP signaling pathway as one of the crucial upstream regulators of Dex induced actin cytoskeletal reorganization and cell adhesion in TM cells, opening an opportunity to target the GPC4/Wnt/PCP pathway for treatment of ocular hypertension in glaucoma.


Actins , Cytoskeletal Proteins , Cytoskeleton , Dexamethasone , Glucocorticoids , Glypicans , Trabecular Meshwork , Humans , Actins/metabolism , Cells, Cultured , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dexamethasone/pharmacology , Glaucoma/metabolism , Glaucoma/pathology , Glucocorticoids/pharmacology , Glypicans/deficiency , Glypicans/metabolism , Intraocular Pressure , Trabecular Meshwork/cytology , Trabecular Meshwork/drug effects , Trabecular Meshwork/metabolism , Wnt Signaling Pathway/drug effects , Cytoskeleton/metabolism , Cell Polarity/drug effects , rho-Associated Kinases/metabolism , Stress Fibers/drug effects , Cell Adhesion/drug effects
2.
Brain ; 144(10): 3226-3238, 2021 11 29.
Article En | MEDLINE | ID: mdl-33964142

Axonal degeneration is an early and ongoing event that causes disability and disease progression in many neurodegenerative disorders of the peripheral and central nervous systems. Chemotherapy-induced peripheral neuropathy (CIPN) is a major cause of morbidity and the main cause of dose reductions and discontinuations in cancer treatment. Preclinical evidence indicates that activation of the Wallerian-like degeneration pathway driven by sterile alpha and TIR motif containing 1 (SARM1) is responsible for axonopathy in CIPN. SARM1 is the central driver of an evolutionarily conserved programme of axonal degeneration downstream of chemical, inflammatory, mechanical or metabolic insults to the axon. SARM1 contains an intrinsic NADase enzymatic activity essential for its pro-degenerative functions, making it a compelling therapeutic target to treat neurodegeneration characterized by axonopathies of the peripheral and central nervous systems. Small molecule SARM1 inhibitors have the potential to prevent axonal degeneration in peripheral and central axonopathies and to provide a transformational disease-modifying treatment for these disorders. Using a biochemical assay for SARM1 NADase we identified a novel series of potent and selective irreversible isothiazole inhibitors of SARM1 enzymatic activity that protected rodent and human axons in vitro. In sciatic nerve axotomy, we observed that these irreversible SARM1 inhibitors decreased a rise in nerve cADPR and plasma neurofilament light chain released from injured sciatic nerves in vivo. In a mouse paclitaxel model of CIPN we determined that Sarm1 knockout mice prevented loss of axonal function, assessed by sensory nerve action potential amplitudes of the tail nerve, in a gene-dosage-dependent manner. In that CIPN model, the irreversible SARM1 inhibitors prevented loss of intraepidermal nerve fibres induced by paclitaxel and provided partial protection of axonal function assessed by sensory nerve action potential amplitude and mechanical allodynia.


Armadillo Domain Proteins/antagonists & inhibitors , Axons/drug effects , Cytoskeletal Proteins/antagonists & inhibitors , Paclitaxel/toxicity , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Thiazoles/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/toxicity , Armadillo Domain Proteins/deficiency , Armadillo Domain Proteins/genetics , Axons/metabolism , Cells, Cultured , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Knockout , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Thiazoles/pharmacology
3.
Am J Med Genet A ; 185(10): 3074-3082, 2021 10.
Article En | MEDLINE | ID: mdl-34047005

PSTPIP1-associated myeloid-related proteinemia inflammatory (PAMI) syndrome caused by mutations in PSTPIP1 is a rare inflammatory disorder that can be easily misdiagnosed. It is characterized by anemia, arthritis, cutaneous inflammation, recurrent infections, growth failure, hepatosplenomegaly, lymphadenopathy, hyperzincemia/hypercalprotectinemia, neutropenia, thrombocytopenia, and elevated inflammatory indicators. This study describes the cases of two pediatric female patients with long-standing recurrent arthralgia in different parts of the extremities and severe anemia, respectively, who were misdiagnosed and treated for aseptic necrosis of the femoral head and severe autoimmune hemolytic anemia, respectively. High-throughput sequencing analysis revealed a de novo heterozygous missense mutation (c.748G > A, p. Glu250Lys) in exon 11 of PSTPIP1 (NM_003978.5) in both patients, which supported a diagnosis of PAMI. The patients were treated with prednisone and etanercept, which improved their symptoms, but neutropenia remained unchanged. These cases highlight the importance of genetic assessment for the accurate diagnosis of PAMI and to ensure adequate and timely treatment of these patients.


Adaptor Proteins, Signal Transducing/genetics , Anemia/diagnosis , Cytoskeletal Proteins/genetics , Inflammation/diagnosis , Neutropenia/diagnosis , Adaptor Proteins, Signal Transducing/deficiency , Anemia/complications , Anemia/genetics , Anemia/pathology , Arthritis/complications , Arthritis/diagnosis , Arthritis/genetics , Arthritis/pathology , Child , Child, Preschool , Cytoskeletal Proteins/deficiency , Diagnostic Errors/prevention & control , Female , Heterozygote , Humans , Inflammation/complications , Inflammation/genetics , Inflammation/pathology , Metal Metabolism, Inborn Errors , Mutation/genetics , Myeloid Cells/pathology , Neutropenia/complications , Neutropenia/genetics , Neutropenia/pathology , Phenotype
4.
Acta Neuropathol Commun ; 9(1): 89, 2021 05 17.
Article En | MEDLINE | ID: mdl-34001261

Traumatic brain injury (TBI) causes chronic symptoms and increased risk of neurodegeneration. Axons in white matter tracts, such as the corpus callosum (CC), are critical components of neural circuits and particularly vulnerable to TBI. Treatments are needed to protect axons from traumatic injury and mitigate post-traumatic neurodegeneration. SARM1 protein is a central driver of axon degeneration through a conserved molecular pathway. Sarm1-/- mice with knockout (KO) of the Sarm1 gene enable genetic proof-of-concept testing of the SARM1 pathway as a therapeutic target. We evaluated Sarm1 deletion effects after TBI using a concussive model that causes traumatic axonal injury and progresses to CC atrophy at 10 weeks, indicating post-traumatic neurodegeneration. Sarm1 wild-type (WT) mice developed significant CC atrophy that was reduced in Sarm1 KO mice. Ultrastructural classification of pathology of individual axons, using electron microscopy, demonstrated that Sarm1 KO preserved more intact axons and reduced damaged or demyelinated axons. Longitudinal MRI studies in live mice identified significantly reduced CC volume after TBI in Sarm1 WT mice that was attenuated in Sarm1 KO mice. MR diffusion tensor imaging detected reduced fractional anisotropy in both genotypes while axial diffusivity remained higher in Sarm1 KO mice. Immunohistochemistry revealed significant attenuation of CC atrophy, myelin loss, and neuroinflammation in Sarm1 KO mice after TBI. Functionally, Sarm1 KO mice exhibited beneficial effects in motor learning and sleep behavior. Based on these findings, Sarm1 inactivation can protect axons and white matter tracts to improve translational outcomes associated with CC atrophy and post-traumatic neurodegeneration.


Armadillo Domain Proteins/deficiency , Axons/metabolism , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/metabolism , Cytoskeletal Proteins/deficiency , Diffusion Tensor Imaging/methods , Gene Silencing/physiology , Animals , Armadillo Domain Proteins/genetics , Axons/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Cytoskeletal Proteins/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/diagnostic imaging , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Treatment Outcome
5.
Neuron ; 109(10): 1636-1656.e8, 2021 05 19.
Article En | MEDLINE | ID: mdl-33831348

Ample evidence indicates that individuals with intellectual disability (ID) are at increased risk of developing stress-related behavioral problems and mood disorders, yet a mechanistic explanation for such a link remains largely elusive. Here, we focused on characterizing the syndromic ID gene oligophrenin-1 (OPHN1). We find that Ophn1 deficiency in mice markedly enhances helpless/depressive-like behavior in the face of repeated/uncontrollable stress. Strikingly, Ophn1 deletion exclusively in parvalbumin (PV) interneurons in the prelimbic medial prefrontal cortex (PL-mPFC) is sufficient to induce helplessness. This behavioral phenotype is mediated by a diminished excitatory drive onto Ophn1-deficient PL-mPFC PV interneurons, leading to hyperactivity in this region. Importantly, suppressing neuronal activity or RhoA/Rho-kinase signaling in the PL-mPFC reverses helpless behavior. Our results identify OPHN1 as a critical regulator of adaptive behavioral responses to stress and shed light onto the mechanistic links among OPHN1 genetic deficits, mPFC circuit dysfunction, and abnormalities in stress-related behaviors.


Cytoskeletal Proteins/metabolism , GTPase-Activating Proteins/metabolism , Interneurons/metabolism , Prefrontal Cortex/metabolism , Stress, Psychological/metabolism , Animals , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , HEK293 Cells , Helplessness, Learned , Humans , Interneurons/physiology , Mice , Mice, Inbred C57BL , Parvalbumins/genetics , Parvalbumins/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Stress, Psychological/physiopathology , Synaptic Transmission
6.
Protein Cell ; 12(8): 621-638, 2021 08.
Article En | MEDLINE | ID: mdl-33871822

Axonal degeneration is one of the key features of neurodegenerative disorders. In the canonical view, axonal degeneration destructs neural connections and promotes detrimental disease defects. Here, we assessed the enteric nervous system (ENS) of the mouse, non-human primate, and human by advanced 3D imaging. We observed the profound neurodegeneration of catecholaminergic axons in human colons with ulcerative colitis, and similarly, in mouse colons during acute dextran sulfate sodium-induced colitis. However, we unexpectedly revealed that blockage of such axonal degeneration by the Sarm1 deletion in mice exacerbated the colitis condition. In contrast, pharmacologic ablation or chemogenetic inhibition of catecholaminergic axons suppressed the colon inflammation. We further showed that the catecholaminergic neurotransmitter norepinephrine exerted a pro-inflammatory function by enhancing the expression of IL-17 cytokines. Together, this study demonstrated that Sarm1-mediated neurodegeneration within the ENS mitigated local inflammation of the colon, uncovering a previously-unrecognized beneficial role of axonal degeneration in this disease context.


Armadillo Domain Proteins/genetics , Colitis, Ulcerative/genetics , Cytoskeletal Proteins/genetics , Enteric Nervous System/metabolism , Neurodegenerative Diseases/genetics , Animals , Armadillo Domain Proteins/deficiency , Catecholamines/metabolism , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/diagnostic imaging , Colitis, Ulcerative/metabolism , Colon/diagnostic imaging , Colon/metabolism , Colon/pathology , Cytoskeletal Proteins/deficiency , Dextran Sulfate/administration & dosage , Disease Models, Animal , Enteric Nervous System/diagnostic imaging , Enteric Nervous System/pathology , Gene Expression Regulation , Humans , Imaging, Three-Dimensional , Interleukin-17/genetics , Interleukin-17/metabolism , Macaca mulatta , Male , Mice , Mice, Knockout , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Neurons/pathology , Norepinephrine/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Neurobiol Dis ; 155: 105368, 2021 07.
Article En | MEDLINE | ID: mdl-33892050

Parkinson's disease (PD) is the most common form of neurodegenerative movement disorder, associated with profound loss of dopaminergic neurons from the basal ganglia. Though loss of dopaminergic neuron cell bodies from the substantia nigra pars compacta is a well-studied feature, atrophy and loss of their axons within the nigrostriatal tract is also emerging as an early event in disease progression. Genes that drive the Wallerian degeneration, like Sterile alpha and toll/interleukin-1 receptor motif containing (Sarm1), are excellent candidates for driving this axon degeneration, given similarities in the morphology of axon degeneration after axotomy and in PD. In the present study we assessed whether Sarm1 contributes to loss of dopaminergic projections in mouse models of PD. In Sarm1 deficient mice, we observed a significant delay in the degeneration of severed dopaminergic axons distal to a 6-OHDA lesion of the medial forebrain bundle (MFB) in the nigrostriatal tract, and an accompanying rescue of morphological, biochemical and behavioural phenotypes. However, we observed no difference compared to controls when striatal terminals were lesioned with 6-OHDA to induce a dying back form of neurodegeneration. Likewise, when PD phenotypes were induced using AAV-induced alpha-synuclein overexpression, we observed similar modest loss of dopaminergic terminals in Sarm1 knockouts and controls. Our data argues that axon degeneration after MFB lesion is Sarm1-dependent, but that other models for PD do not require Sarm1, or that Sarm1 acts with other redundant genetic pathways. This work adds to a growing body of evidence indicating Sarm1 contributes to some, but not all types of neurodegeneration, and supports the notion that while axon degeneration in many context appears morphologically similar, a diversity of axon degeneration programs exist.


Armadillo Domain Proteins/genetics , Axons/pathology , Cytoskeletal Proteins/genetics , Genetic Variation/physiology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Animals , Armadillo Domain Proteins/deficiency , Axons/metabolism , Cytoskeletal Proteins/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced
8.
Exp Neurol ; 339: 113636, 2021 05.
Article En | MEDLINE | ID: mdl-33548217

Axonal degeneration contributes to the pathogenesis of many neurodegenerative disorders, motivating efforts to dissect the mechanism of pathological axon loss in order to develop therapies for axonal preservation. SARM1 is a particularly attractive therapeutic target, as it is an inducible NAD+ cleaving enzyme that is required for axon loss in multiple mouse models of traumatic and degenerative neurological disease. However, it is essential to establish whether SARM1 triggers axon degeneration in human neurons before proceeding with the development of SARM1-directed therapeutics. Here we combine genome engineering with the production of human stem cell-derived neurons to test the role of human SARM1 in traumatic and neurotoxic axon degeneration. We have generated two independent SARM1 knockout human iPSC lines that do not express SARM1 protein upon differentiation into neurons. We have developed a modified sensory neuron differentiation protocol that generates human sensory neurons with high yield and purity. We find that SARM1 is required for axon degeneration in response to both physical trauma and in a cellular model of chemotherapy-induced peripheral neuropathy. Finally, we identify cADPR as a biomarker of SARM1 enzyme activity in both healthy and injured human sensory neurons. These findings are consistent with prior molecular and cellular studies in mouse neurons, and highlight the therapeutic potential of SARM1 inhibition for the prevention and treatment of human neurological disease.


Armadillo Domain Proteins/deficiency , Armadillo Domain Proteins/genetics , Axons/metabolism , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Sensory Receptor Cells/metabolism , Base Sequence , Cell Line , Gene Knockout Techniques/methods , Humans , Induced Pluripotent Stem Cells/metabolism
9.
Semin Cell Dev Biol ; 110: 104-112, 2021 02.
Article En | MEDLINE | ID: mdl-32693941

How developing and growing organisms attain their proper shape is a central problem of developmental biology. In this review, we investigate this question with respect to how the body axis and spine form in their characteristic linear head-to-tail fashion in vertebrates. Recent work in the zebrafish has implicated motile cilia and cerebrospinal fluid flow in axial morphogenesis and spinal straightness. We begin by introducing motile cilia, the fluid flows they generate and their roles in zebrafish development and growth. We then describe how cilia control body and spine shape through sensory cells in the spinal canal, a thread-like extracellular structure called the Reissner fiber, and expression of neuropeptide signals. Last, we discuss zebrafish mutants in which spinal straightness breaks down and three-dimensional curves form. These curves resemble the common but little-understood human disease Idiopathic Scoliosis. Zebrafish research is therefore poised to make progress in our understanding of this condition and, more generally, how body and spine shape is acquired and maintained through development and growth.


Cell Adhesion Molecules, Neuronal/genetics , Cilia/metabolism , Cytoskeletal Proteins/genetics , Morphogenesis/genetics , Scoliosis/genetics , Spine/metabolism , Zebrafish Proteins/genetics , Animals , Axoneme/metabolism , Axoneme/ultrastructure , Cell Adhesion Molecules, Neuronal/deficiency , Cerebrospinal Fluid/chemistry , Cilia/pathology , Cilia/ultrastructure , Cytoskeletal Proteins/deficiency , Disease Models, Animal , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Humans , Mutation , Scoliosis/metabolism , Scoliosis/pathology , Signal Transduction , Spine/abnormalities , Spine/growth & development , Urotensins/genetics , Zebrafish , Zebrafish Proteins/deficiency
10.
Am J Med Genet A ; 185(3): 877-883, 2021 03.
Article En | MEDLINE | ID: mdl-33346930

Disruption of the autism susceptibility candidate 2 (AUTS2) gene through genomic rearrangements, copy number variations (CNVs), and intragenic deletions and mutations, has been recurrently involved in syndromic forms of developmental delay and intellectual disability, known as AUTS2 syndrome. The AUTS2 gene plays an important role in regulation of neuronal migration, and when altered, associates with a variable phenotype from severely to mildly affected patients. The more severe phenotypes significantly correlate with the presence of defects affecting the C-terminus part of the gene. This article reports a new patient with a syndromic neurodevelopmental disorder, who presents a deletion of 30 nucleotides in the exon 9 of the AUTS2 gene. Importantly, this deletion includes the transcription start site for the AUTS2 short transcript isoform, which has an important role in brain development. Gene expression analysis of AUTS2 full-length and short isoforms revealed that the deletion found in this patient causes a remarkable reduction in the expression level, not only of the short isoform, but also of the full AUTS2 transcripts. This report adds more evidence for the role of mutated AUTS2 short transcripts in the development of a severe phenotype in the AUTS2 syndrome.


Cytoskeletal Proteins/genetics , Exons/genetics , Neurodevelopmental Disorders/genetics , Sequence Deletion , Transcription Factors/genetics , Transcription Initiation Site , Child, Preschool , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/deficiency , Dwarfism/genetics , Gene Expression Regulation , Genetic Association Studies , Humans , Male , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Syndrome , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transcription, Genetic
11.
Clin Genet ; 99(4): 540-546, 2021 04.
Article En | MEDLINE | ID: mdl-33372278

Biallelic pathogenic variants in POC1A result in SOFT (Short-stature, Onychodysplasia, Facial-dysmorphism, and hypoTrichosis) and variant POC1A-related (vPOC1A) syndromes. The latter, nowadays described in only two unrelated subjects, is associated with a restricted spectrum of variants falling in exon 10, which is naturally skipped in a specific POC1A mRNA. The synthesis of an amount of a POC1A isoform from this transcript in individuals with vPOC1A syndrome has been believed as the likely explanation for such a genotype-phenotype correlation. Here, we illustrate the clinical and molecular findings in a woman who resulted to be compound heterozygous for a recurrent frameshift variant in exon 10 and a novel variant in exon 9 of POC1A. Phenotypic characteristics of this woman included severe hyperinsulinemic dyslipidemia, acanthosis nigricans, moderate growth restriction, and dysmorphisms. These manifestations overlap the clinical features of the two previously published individuals with vPOC1A syndrome. RT-PCR analysis on peripheral blood and subsequent sequencing of the obtained amplicons demonstrated a variety of POC1A alternative transcripts that resulted to be expressed in the proband, in the healthy mother, and in controls. We illustrate the possible consequences of the two POC1A identified variants in an attempt to explain pleiotropy in vPOC1A syndrome.


Cell Cycle Proteins/genetics , Congenital Hyperinsulinism/genetics , Cytoskeletal Proteins/genetics , Dyslipidemias/genetics , Acanthosis Nigricans/genetics , Adult , Age of Onset , Cell Cycle Proteins/deficiency , Computer Simulation , Congenital Hyperinsulinism/drug therapy , Cytoskeletal Proteins/deficiency , DNA, Complementary/genetics , Dyslipidemias/drug therapy , Exons/genetics , Fatty Acids, Unsaturated/therapeutic use , Female , Frameshift Mutation , Heterozygote , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Insulin Resistance , Metformin/therapeutic use , Middle Aged , Pedigree , Phenotype , Plasmapheresis , Protein Isoforms/genetics , Syndrome , Transcription, Genetic
12.
Nat Commun ; 11(1): 5520, 2020 11 02.
Article En | MEDLINE | ID: mdl-33139725

Axonemal dynein ATPases direct ciliary and flagellar beating via adenosine triphosphate (ATP) hydrolysis. The modulatory effect of adenosine monophosphate (AMP) and adenosine diphosphate (ADP) on flagellar beating is not fully understood. Here, we describe a deficiency of cilia and flagella associated protein 45 (CFAP45) in humans and mice that presents a motile ciliopathy featuring situs inversus totalis and asthenospermia. CFAP45-deficient cilia and flagella show normal morphology and axonemal ultrastructure. Proteomic profiling links CFAP45 to an axonemal module including dynein ATPases and adenylate kinase as well as CFAP52, whose mutations cause a similar ciliopathy. CFAP45 binds AMP in vitro, consistent with structural modelling that identifies an AMP-binding interface between CFAP45 and AK8. Microtubule sliding of dyskinetic sperm from Cfap45-/- mice is rescued with the addition of either AMP or ADP with ATP, compared to ATP alone. We propose that CFAP45 supports mammalian ciliary and flagellar beating via an adenine nucleotide homeostasis module.


Adenine Nucleotides/metabolism , Asthenozoospermia/genetics , Cytoskeletal Proteins/deficiency , Situs Inversus/genetics , Adolescent , Adult , Animals , Asthenozoospermia/pathology , Axoneme/ultrastructure , CRISPR-Cas Systems/genetics , Cilia/metabolism , Cilia/ultrastructure , Cytoskeletal Proteins/genetics , DNA Mutational Analysis , Disease Models, Animal , Epididymis/pathology , Female , Flagella/metabolism , Flagella/ultrastructure , Humans , Loss of Function Mutation , Male , Mice , Mice, Knockout , Middle Aged , Planarians/cytology , Planarians/genetics , Planarians/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/pathology , Situs Inversus/diagnostic imaging , Situs Inversus/pathology , Sperm Motility/genetics , Tomography, X-Ray Computed , Exome Sequencing
13.
Sci Rep ; 10(1): 16947, 2020 10 12.
Article En | MEDLINE | ID: mdl-33046791

Protein 4.1N, a member of the protein 4.1 family, is highly expressed in the brain. But its function remains to be fully defined. Using 4.1N-/- mice, we explored the function of 4.1N in vivo. We show that 4.1N-/- mice were born at a significantly reduced Mendelian ratio and exhibited high mortality between 3 to 5 weeks of age. Live 4.1N-/- mice were smaller than 4.1N+/+ mice. Notably, while there were no significant differences in organ/body weight ratio for most of the organs, the testis/body and ovary/body ratio were dramatically decreased in 4.1N-/- mice, demonstrating selective effects of 4.1N deficiency on the development of the reproductive systems. Histopathology of the reproductive organs showed atrophy of both testis and ovary. Specifically, in the testis there is a lack of spermatogenesis, lack of leydig cells and lack of mature sperm. Similarly, in the ovary there is a lack of follicular development and lack of corpora lutea formation, as well as lack of secretory changes in the endometrium. Examination of pituitary glands revealed that the secretory granules were significantly decreased in pituitary glands of 4.1N-/- compared to 4.1N+/+. Moreover, while GnRH was expressed in both neuronal cell body and axons in the hypothalamus of 4.1N+/+ mice, it was only expressed in the cell body but not the axons of 4.1N-/- mice. Our findings uncover a novel role for 4.1N in the axis of hypothalamus-pituitary gland-reproductive system.


Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/physiology , Genitalia/metabolism , Genitalia/pathology , Membrane Proteins/deficiency , Membrane Proteins/physiology , Neuropeptides/deficiency , Neuropeptides/physiology , Neurosecretory Systems/metabolism , Neurosecretory Systems/pathology , Animals , Cytoskeletal Proteins/genetics , Female , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Male , Membrane Proteins/genetics , Mice, Knockout , Neuropeptides/genetics , Organ Size , Ovary/pathology , Pituitary Gland/metabolism , Pituitary Gland/pathology , Spermatogenesis/genetics , Testis/pathology
14.
Proc Natl Acad Sci U S A ; 117(39): 24326-24335, 2020 09 29.
Article En | MEDLINE | ID: mdl-32929022

Kindlin-3 (K3)-mediated integrin adhesion controls homing and bone marrow (BM) retention of normal hematopoietic cells. However, the role of K3 in leukemic stem cell (LSC) retention and growth in the remodeled tumor-promoting BM is unclear. We report that loss of K3 in a mouse model of chronic myeloid leukemia (CML) triggers the release of LSCs from the BM into the circulation and impairs their retention, proliferation, and survival in secondary organs, which curbs CML development, progression, and metastatic dissemination. We found de novo expression of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) on CML-LSCs but not normal hematopoietic stem cells and this enabled us to specifically deplete K3 with a CTLA-4-binding RNA aptamer linked to a K3-siRNA (small interfering RNA) in CTLA-4+ LSCs in vivo, which mobilized LSCs in the BM, induced disease remission, and prolonged survival of mice with CML. Thus, disrupting interactions of LSCs with the BM environment is a promising strategy to halt the disease-inducing and relapse potential of LSCs.


Bone Marrow/metabolism , Cytoskeletal Proteins/deficiency , Leukemia, Myeloid/metabolism , Neoplastic Stem Cells/metabolism , Animals , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Cell Movement , Cytoskeletal Proteins/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplastic Stem Cells/cytology , Stem Cell Niche , Tumor Microenvironment
15.
Mol Vis ; 26: 563-573, 2020.
Article En | MEDLINE | ID: mdl-32818018

Purpose: In a previous study, we identified the Asn450Tyr mutant myocilin gene (Myoc-N450Y) in the pedigree of families with juvenile open angle glaucoma (JOAG), but whether N450Y is a pathogenic mutation remained to be determined. The present study aimed at exploring the role of Myoc-N450Y in primary human trabecular meshwork (HTM) cells. Methods: Primary HTM cells were infected with lentivirus with wild-type myocilin (Myoc-WT) or Myoc-N450Y. Primary HTM cells overexpressing Myoc-WT or Myoc-N450Y was treated with sodium 4-phenylbutyrate (4-PBA) or not. The secretion and intracellular distribution of Myoc were analyzed with western blotting and immunofluorescence. Expression of endoplasmic reticulum (ER) stress-related proteins was detected with quantitative real-time PCR (qRT-PCR) and western blotting. Cell viability, apoptosis, and expression of the related proteins were examined with Cell Counting Kit-8 (CCK-8), flow cytometry analysis, and western blotting, respectively. Results: We found that non-secretion of Myoc-N450Y induced ER stress by colocalization with the ER marker calreticulin (CALR), and upregulating the expression of ER stress markers in primary HTM cells. Moreover, overexpression of Myoc-N450Y inhibited the viability and induced apoptosis of primary HTM cells, and inhibition of PI3K/AKT signaling was induced by ER stress. Reduction in ER stress with 4-PBA decreased the level of ER stress markers, promoted secretion, and prevented accumulation of myocilin in the Myoc-N450Y group. Apoptosis was rescued, and inhibition of PI3K/AKT signaling was reversed, after PBA treatment in primary HTM cells with Myoc-N450Y overexpression. Conclusions: The study results suggest that Myoc-N450Y promotes apoptosis of primary HTM cells via the ER stress-induced apoptosis pathway, in which the PI3K/AKT signaling pathway plays a crucial role.


Cytoskeletal Proteins/genetics , Eye Proteins/genetics , Glaucoma, Open-Angle/genetics , Glycoproteins/genetics , Mutation , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Trabecular Meshwork/metabolism , Apoptosis/genetics , Aqueous Humor/metabolism , Calreticulin/genetics , Calreticulin/metabolism , Cell Survival , Cytoskeletal Proteins/deficiency , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/pathology , Glycoproteins/deficiency , Humans , Intraocular Pressure , Phenylbutyrates/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology
16.
Front Immunol ; 11: 1559, 2020.
Article En | MEDLINE | ID: mdl-32849529

Activation of NF-κB transcription factors is critical for innate immune cells to induce inflammation and fight against microbial pathogens. On the other hand, the excessive and prolonged activation of NF-κB causes massive inflammatory damage to the host, suggesting that regulatory mechanisms to promptly terminate NF-κB activation are important to prevent immunopathology. We have previously reported that PDLIM2, a PDZ-LIM domain-containing protein, is a nuclear ubiquitin E3 ligase that targets the p65 subunit of NF-κB for degradation, thereby suppressing NF-κB activation. Here we show that PDLIM7, another member of LIM protein family, is also a ubiquitin E3 ligase that inhibits NF-κB-mediated inflammatory responses. PDLIM7 directly polyubiquitinates p65 and promotes its proteasomal degradation. Moreover, PDLIM7 heterodimerizes with PDLIM2 to promote synergistic PDLIM2-mediated degradation of p65. Mechanistically, PDLIM7 promotes K63-linked ubiquitination of PDLIM2 and then the proteasome/autophagosome cargo protein p62/Sqstm1 binds to both polyubiquitinated PDLIM2 and the proteasome, thereby facilitating the delivery of the NF-κB-PDLIM2 complex to the proteasome and subsequent p65 degradation. Consistently, double knockdown of PDLIM7 and either PDLIM2 or p62/Sqstm1 results in augmented proinflammatory cytokine production compared to control cells or single knockdown cells. These data delineate a new role for PDLIM7 and p62/Sqstm1 in the regulation of NF-κB signaling by bridging a ubiquitin E3 ligase and the proteasome.


Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Disease Susceptibility , Inflammation/etiology , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , NF-kappa B/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Enzyme Activation , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/deficiency , LIM Domain Proteins/genetics , Lipopolysaccharides/adverse effects , Lipopolysaccharides/immunology , Mice , NF-kappa B/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Proteolysis , RNA, Small Interfering/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
17.
J Virol ; 94(18)2020 08 31.
Article En | MEDLINE | ID: mdl-32611756

Cellular intrinsic immunity, mediated by the expression of an array of interferon-stimulated antiviral genes, is a vital part of host defense. We have previously used a bioinformatic screen to identify two interferon-stimulated genes (ISG) with poorly characterized function, interferon-induced protein 44 (IFI44) and interferon-induced protein 44-like (IFI44L), as potentially being important in respiratory syncytial virus (RSV) infection. Using overexpression systems, CRISPR-Cas9-mediated knockout, and a knockout mouse model, we investigated the antiviral capability of these genes in the control of RSV replication. Overexpression of IFI44 or IFI44L was sufficient to restrict RSV infection at an early time postinfection. Knocking out these genes in mammalian airway epithelial cells increased levels of infection. Both genes express antiproliferative factors that have no effect on RSV attachment but reduce RSV replication in a minigenome assay. The loss of Ifi44 was associated with a more severe infection phenotype in a mouse model of infection. These studies demonstrate a function for IFI44 and IFI44L in controlling RSV infection.IMPORTANCE RSV infects all children under 2 years of age, but only a subset of children get severe disease. We hypothesize that susceptibility to severe RSV necessitating hospitalization in children without predefined risk factors is, in part, mediated at the antiviral gene level. However, there is a large array of antiviral genes, particularly in the ISG family, the mechanism of which is poorly understood. Having previously identified IFI44 and IFI44L as possible genes of interest in a bioinformatic screen, we dissected the function of these two genes in the control of RSV. Through a range of overexpression and knockout studies, we show that the genes are antiviral and antiproliferative. This study is important because IFI44 and IFI44L are upregulated after a wide range of viral infections, and IFI44L can serve as a diagnostic biomarker of viral infection.


Antigens/immunology , Cytoskeletal Proteins/immunology , Host-Pathogen Interactions/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Tumor Suppressor Proteins/immunology , A549 Cells , Animals , Antigens/genetics , Biological Assay , CRISPR-Cas Systems , Cell Line, Tumor , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Disease Models, Animal , Epithelial Cells , Gene Editing , Gene Expression Regulation , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , Immunity, Innate , Infant , Mice , Mice, Knockout , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Signal Transduction , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Virus Replication
18.
Theranostics ; 10(14): 6182-6200, 2020.
Article En | MEDLINE | ID: mdl-32483447

Rationale: Smooth muscle-motility disorders are mainly characterized by impaired contractility and functional intestinal obstruction. Some of these cases are caused by genetic mutations of smooth muscle genes ACTA2, ACTG2, MYH11, MYLK and LMOD1. Still the etiology is complex and multifactorial and the underlying pathology is poorly understood. Integrin interaction protein Kindlin-2 is widely expressed in striated and smooth muscle cells (SMC). However, the function of Kindlin-2 in the smooth muscle remains elusive. Methods: We generated two mouse models using different cre promoter transgenic mice, Kindlin-2fl/fl SM22α-cre+ (cKO mice) and Kindlin-2fl/fl; MYH-cre+ (iKO mice). Embryos and adult tissues were prepared for hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC) and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) apoptosis assay. We investigated ultrastructure changes of mouse smooth muscle using transmission electron microscopy (TEM) and measured smooth muscle contractile force in mounting aortic and intestinal rings using the multiwire myograph system (DMT 620M). In addition, cell traction force microscopy (CTFM) was applied to observe the functional change of primary SMC after Kindlin-2 depletion by RNAi. Results: Depletion of Kindlin-2 encoding gene Fermt2 in embryonic smooth muscles leads to apoptosis, downregulates the key components of SMC, impairs smooth muscle development, and finally causes embryonic death at E14.5. Tamoxifen-induced Kindlin-2-specific knockout in adult mouse smooth muscle showed decreased blood pressure, intestinal hypoperistalsis, and eventually died of intestinal obstruction. Kindlin-2 depletion also leads to downregulated Myh11, α-SMA, and CNN, shortened myofilament, broken myofibrils, and impaired contractility of the smooth muscles in iKO mice. Mechanistically, loss of Kindlin-2 decreases Ca2+ influx in primary vascular smooth muscle cells (PVSMC) by downregulating the expression of calcium-binding protein S100A14 and STIM1. Conclusion: We demonstrated that Kindlin-2 is essential for maintaining the normal structure and function of smooth muscles. Loss of Kindlin-2 impairs smooth muscle formation during embryonic development by inducing apoptosis and jeopardizes the contraction of adult smooth muscle by blocking Ca2+ influx that leads to intestinal obstruction. Mice with Kindlin-2 depletion in adult smooth muscle could be a potent animal model of intestinal obstruction for disease research, drug treatment and prognosis.


Calcium-Binding Proteins/metabolism , Cytoskeletal Proteins/deficiency , Intestinal Obstruction/pathology , Muscle Proteins/deficiency , Muscle, Smooth/pathology , Animals , Cell Movement , Cytoskeletal Proteins/genetics , Disease Models, Animal , Intestinal Obstruction/etiology , Intestinal Obstruction/metabolism , Mice , Mice, Knockout , Muscle Contraction , Muscle Proteins/genetics , Muscle, Smooth/metabolism
19.
Methods Mol Biol ; 2143: 145-157, 2020.
Article En | MEDLINE | ID: mdl-32524478

We describe here an organotypic culture system we have used to investigate mechanisms that maintain structure and function of axon terminals at the neuromuscular junction (NMJ). We developed this by taking advantage of the slow Wallerian degeneration phenotype in mutant Wlds mice, using these to compare preservation of NMJs with degeneration in nerve-muscle preparations from wild-type mice. We take hind limb tibial nerve/flexor digitorum brevis and lumbrical muscles and incubate them in mammalian physiological saline at 32 °C for 24-48 h. Integrity of NMJs can then be compared using a combination of electrophysiological and morphological techniques. We illustrate our method with data showing synaptic preservation ex vivo in nerve-muscle explants from Sarm-1 null-mutant mice. The ex vivo assays of NMJ integrity we describe here may therefore be useful for detailed investigation of synaptic maintenance and degeneration.


Neuromuscular Junction/physiology , Organ Culture Techniques/methods , Wallerian Degeneration/physiopathology , Animals , Armadillo Domain Proteins/deficiency , Axons/physiology , Cytoskeletal Proteins/deficiency , Dissection/methods , Electrophysiology/methods , Female , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Muscle, Skeletal , Neuromuscular Junction/ultrastructure , Organ Culture Techniques/instrumentation , Synapses/ultrastructure , Tibial Nerve
20.
J Leukoc Biol ; 107(6): 883-892, 2020 06.
Article En | MEDLINE | ID: mdl-32386455

Nonclassical monocytes maintain vascular homeostasis by patrolling the vascular endothelium, responding to inflammatory signals, and scavenging cellular debris. Nonclassical monocytes also prevent metastatic tumor cells from seeding new tissues, but whether the patrolling function of nonclassical monocytes is required for this process is unknown. To answer this question, we utilized an inducible-knockout mouse that exhibits loss of the integrin-adaptor protein Kindlin-3 specifically in nonclassical monocytes. We show that Kindlin-3-deficient nonclassical monocytes are unable to patrol the vascular endothelium in either the lungs or periphery. We also find that Kindlin-3-deficient nonclassical monocytes cannot firmly adhere to, and instead "slip" along, the vascular endothelium. Loss of patrolling activity by nonclassical monocytes was phenocopied by ablation of LFA-1, an integrin-binding partner of Kindlin-3. When B16F10 murine melanoma tumor cells were introduced into Kindlin-3-deficient mice, nonclassical monocytes showed defective patrolling towards tumor cells and failure to ingest tumor particles in vivo. Consequently, we observed a significant, 4-fold increase in lung tumor metastases in mice possessing Kindlin-3-deficient nonclassical monocytes. Thus, we conclude that the patrolling function of nonclassical monocytes is mediated by Kindlin-3 and essential for these cells to maintain vascular endothelial homeostasis and prevent tumor metastasis to the lung.


Cytoskeletal Proteins/genetics , Gene Expression Regulation, Neoplastic , Lymphocyte Function-Associated Antigen-1/genetics , Melanoma, Experimental/genetics , Monocytes/immunology , Phagocytosis , Skin Neoplasms/genetics , Animals , Bone Marrow/immunology , Bone Marrow Transplantation , Cell Adhesion , Cell Communication/immunology , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Female , Humans , Injections, Intravenous , Lung/blood supply , Lung/immunology , Lung/pathology , Lymphocyte Function-Associated Antigen-1/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Mice , Mice, Knockout , Monocytes/pathology , Neoplastic Cells, Circulating/immunology , Neoplastic Cells, Circulating/pathology , Primary Cell Culture , Signal Transduction , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Whole-Body Irradiation
...