Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 735
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 33(13): 1107-1119, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38507070

ABSTRACT

The dystrophin-glycoprotein complex (DGC) plays a crucial role in maintaining the structural integrity of the plasma membrane and the neuromuscular junction. In this study, we investigated the impact of the deficiency of α-dystrobrevin (αdbn), a component of the DGC, on the homeostasis of intracellular organelles, specifically mitochondria and the sarcoplasmic reticulum (SR). In αdbn deficient muscles, we observed a significant increase in the membrane-bound ATP synthase complex levels, a marker for mitochondria in oxidative muscle fiber types compared to wild-type. Furthermore, examination of muscle fibers deficient in αdbn using electron microscopy revealed profound alterations in the organization of mitochondria and the SR within certain myofibrils of muscle fibers. This included the formation of hyper-branched intermyofibrillar mitochondria with extended connections, an extensive network spanning several myofibrils, and a substantial increase in the number/density of subsarcolemmal mitochondria. Concurrently, in some cases, we observed significant structural alterations in mitochondria, such as cristae loss, fragmentation, swelling, and the formation of vacuoles and inclusions within the mitochondrial matrix cristae. Muscles deficient in αdbn also displayed notable alterations in the morphology of the SR, along with the formation of distinct anomalous concentric SR structures known as whorls. These whorls were prevalent in αdbn-deficient mice but were absent in wild-type muscles. These results suggest a crucial role of the DGC αdbn in regulating intracellular organelles, particularly mitochondria and the SR, within muscle cells. The remodeling of the SR and the formation of whorls may represent a novel mechanism of the unfolded protein response (UPR) in muscle cells.


Subject(s)
Dystrophin-Associated Proteins , Dystrophin , Mitochondria , Sarcoplasmic Reticulum , Animals , Mice , Dystrophin/genetics , Dystrophin/metabolism , Dystrophin/deficiency , Dystrophin-Associated Proteins/genetics , Dystrophin-Associated Proteins/metabolism , Glycoproteins/metabolism , Glycoproteins/genetics , Glycoproteins/deficiency , Mice, Knockout , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondria/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myofibrils/metabolism , Myofibrils/ultrastructure , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/ultrastructure
2.
J Virol ; 97(10): e0083223, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37796128

ABSTRACT

IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of several B cell malignancies and Kaposi's sarcoma. We analyzed the function of K8.1, the major antigenic component of the KSHV virion in the infection of different cells. To do this, we deleted K8.1 from the viral genome. It was found that K8.1 is critical for the infection of certain epithelial cells, e.g., a skin model cell line but not for infection of many other cells. K8.1 was found to mediate attachment of the virus to cells where it plays a role in infection. In contrast, we did not find K8.1 or a related protein from a closely related monkey virus to activate fusion of the viral and cellular membranes, at least not under the conditions tested. These findings suggest that K8.1 functions in a highly cell-specific manner during KSHV entry, playing a crucial role in the attachment of KSHV to, e.g., skin epithelial cells.


Subject(s)
Glycoproteins , Herpesvirus 8, Human , Keratinocytes , Viral Proteins , Virus Attachment , Virus Internalization , Humans , Glycoproteins/deficiency , Glycoproteins/genetics , Glycoproteins/metabolism , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/physiology , Keratinocytes/metabolism , Keratinocytes/virology , Sarcoma, Kaposi/virology , Viral Proteins/genetics , Viral Proteins/metabolism , Membrane Fusion , Skin/cytology
3.
Nature ; 547(7662): 227-231, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28581498

ABSTRACT

The regenerative capacity of the adult mammalian heart is limited, because of the reduced ability of cardiomyocytes to progress through mitosis. Endogenous cardiomyocytes have regenerative capacity at birth but this capacity is lost postnatally, with subsequent organ growth occurring through cardiomyocyte hypertrophy. The Hippo pathway, a conserved kinase cascade, inhibits cardiomyocyte proliferation in the developing heart to control heart size and prevents regeneration in the adult heart. The dystrophin-glycoprotein complex (DGC), a multicomponent transmembrane complex linking the actin cytoskeleton to extracellular matrix, is essential for cardiomyocyte homeostasis. DGC deficiency in humans results in muscular dystrophy, including the lethal Duchenne muscular dystrophy. Here we show that the DGC component dystroglycan 1 (Dag1) directly binds to the Hippo pathway effector Yap to inhibit cardiomyocyte proliferation in mice. The Yap-Dag1 interaction was enhanced by Hippo-induced Yap phosphorylation, revealing a connection between Hippo pathway function and the DGC. After injury, Hippo-deficient postnatal mouse hearts maintained organ size control by repairing the defect with correct dimensions, whereas postnatal hearts deficient in both Hippo and the DGC showed cardiomyocyte overproliferation at the injury site. In the hearts of mature Mdx mice (which have a point mutation in Dmd)-a model of Duchenne muscular dystrophy-Hippo deficiency protected against overload-induced heart failure.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dystrophin/metabolism , Glycoproteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Myocytes, Cardiac/cytology , Phosphoproteins/metabolism , Animals , Cardiomyopathies , Cell Cycle Proteins , Cell Proliferation , Dystroglycans/metabolism , Dystrophin/deficiency , Dystrophin/genetics , Glycoproteins/deficiency , Heart Failure/genetics , Heart Failure/prevention & control , Hippo Signaling Pathway , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Multiprotein Complexes/deficiency , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Myocytes, Cardiac/metabolism , Organ Size , Phosphorylation , Pressure , Protein Binding , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , YAP-Signaling Proteins
4.
Nature ; 540(7631): 124-128, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27819681

ABSTRACT

Receptor-interacting protein kinase 1 (RIPK1) regulates cell death and inflammation through kinase-dependent and -independent functions. RIPK1 kinase activity induces caspase-8-dependent apoptosis and RIPK3 and mixed lineage kinase like (MLKL)-dependent necroptosis. In addition, RIPK1 inhibits apoptosis and necroptosis through kinase-independent functions, which are important for late embryonic development and the prevention of inflammation in epithelial barriers. The mechanism by which RIPK1 counteracts RIPK3-MLKL-mediated necroptosis has remained unknown. Here we show that RIPK1 prevents skin inflammation by inhibiting activation of RIPK3-MLKL-dependent necroptosis mediated by Z-DNA binding protein 1 (ZBP1, also known as DAI or DLM1). ZBP1 deficiency inhibited keratinocyte necroptosis and skin inflammation in mice with epidermis-specific RIPK1 knockout. Moreover, mutation of the conserved RIP homotypic interaction motif (RHIM) of endogenous mouse RIPK1 (RIPK1mRHIM) caused perinatal lethality that was prevented by RIPK3, MLKL or ZBP1 deficiency. Furthermore, mice expressing only RIPK1mRHIM in keratinocytes developed skin inflammation that was abrogated by MLKL or ZBP1 deficiency. Mechanistically, ZBP1 interacted strongly with phosphorylated RIPK3 in cells expressing RIPK1mRHIM, suggesting that the RIPK1 RHIM prevents ZBP1 from binding and activating RIPK3. Collectively, these results show that RIPK1 prevents perinatal death as well as skin inflammation in adult mice by inhibiting ZBP1-induced necroptosis. Furthermore, these findings identify ZBP1 as a critical mediator of inflammation beyond its previously known role in antiviral defence and suggest that ZBP1 might be implicated in the pathogenesis of necroptosis-associated inflammatory diseases.


Subject(s)
Apoptosis , Glycoproteins/antagonists & inhibitors , Glycoproteins/metabolism , Inflammation/metabolism , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Glycoproteins/deficiency , Inflammation/genetics , Inflammation/pathology , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mutation , Phosphorylation , Protein Domains/genetics , Protein Kinases/deficiency , Protein Kinases/metabolism , RNA-Binding Proteins , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Skin/metabolism , Skin/pathology
5.
Nature ; 540(7631): 129-133, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27819682

ABSTRACT

Receptor-interacting protein kinase 1 (RIPK1) promotes cell survival-mice lacking RIPK1 die perinatally, exhibiting aberrant caspase-8-dependent apoptosis and mixed lineage kinase-like (MLKL)-dependent necroptosis. However, mice expressing catalytically inactive RIPK1 are viable, and an ill-defined pro-survival function for the RIPK1 scaffold has therefore been proposed. Here we show that the RIP homotypic interaction motif (RHIM) in RIPK1 prevents the RHIM-containing adaptor protein ZBP1 (Z-DNA binding protein 1; also known as DAI or DLM1) from activating RIPK3 upstream of MLKL. Ripk1RHIM/RHIM mice that expressed mutant RIPK1 with critical RHIM residues IQIG mutated to AAAA died around birth and exhibited RIPK3 autophosphorylation on Thr231 and Ser232, which is a hallmark of necroptosis, in the skin and thymus. Blocking necroptosis with catalytically inactive RIPK3(D161N), RHIM mutant RIPK3, RIPK3 deficiency, or MLKL deficiency prevented lethality in Ripk1RHIM/RHIM mice. Loss of ZBP1, which engages RIPK3 in response to certain viruses but previously had no defined role in development, also prevented perinatal lethality in Ripk1RHIM/RHIM mice. Consistent with the RHIM of RIPK1 functioning as a brake that prevents ZBP1 from engaging the RIPK3 RHIM, ZBP1 interacted with RIPK3 in Ripk1RHIM/RHIMMlkl-/- macrophages, but not in wild-type, Mlkl-/- or Ripk1RHIM/RHIMRipk3RHIM/RHIM macrophages. Collectively, these findings indicate that the RHIM of RIPK1 is critical for preventing ZBP1/RIPK3/MLKL-dependent necroptosis during development.


Subject(s)
Apoptosis , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Glycoproteins/antagonists & inhibitors , Glycoproteins/metabolism , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/metabolism , Amino Acid Motifs , Animals , Animals, Newborn , Caspase 8/genetics , Caspase 8/metabolism , Embryo, Mammalian/cytology , Female , Glycoproteins/chemistry , Glycoproteins/deficiency , Macrophages/metabolism , Male , Mice , Mutation , Phosphorylation , Protein Binding , Protein Kinases/deficiency , Protein Kinases/metabolism , RNA-Binding Proteins , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/pharmacology
6.
Development ; 145(2)2018 01 17.
Article in English | MEDLINE | ID: mdl-29343636

ABSTRACT

During development, precerebellar neurons migrate tangentially from the dorsal hindbrain to the floor plate. Their axons cross it but their cell bodies stop their ventral migration upon reaching the midline. It has previously been shown that Slit chemorepellents and their receptors, Robo1 and Robo2, might control the migration of precerebellar neurons in a repulsive manner. Here, we have used a conditional knockout strategy in mice to test this hypothesis. We show that the targeted inactivation of the expression of Robo1 and Robo2 receptors in precerebellar neurons does not perturb their migration and that they still stop at the midline. The selective ablation of the expression of all three Slit proteins in floor-plate cells has no effect on pontine neurons and only induces the migration of a small subset of inferior olivary neurons across the floor plate. Likewise, we show that the expression of Slit proteins in the facial nucleus is dispensable for pontine neuron migration. Together, these results show that Robo1 and Robo2 receptors act non-cell autonomously in migrating precerebellar neurons and that floor-plate signals, other than Slit proteins, must exist to prevent midline crossing.


Subject(s)
Cell Movement/physiology , Cerebellum/embryology , Glycoproteins/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Receptors, Immunologic/physiology , Animals , Cerebellum/cytology , Female , Glycoproteins/deficiency , Glycoproteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurogenesis/physiology , Pregnancy , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Signal Transduction , Roundabout Proteins
8.
J Mol Cell Cardiol ; 142: 39-52, 2020 05.
Article in English | MEDLINE | ID: mdl-32268148

ABSTRACT

Vascular calcification is a pathological process closely related to atherosclerosis, diabetic vascular diseases, vascular injury, hypertension, chronic kidney disease and aging. Lethal giant larvae 1 (LGL1) is known as a key regulator of cell polarity and plays an important role in tumorigenesis. However, whether LGL1 regulates vascular calcification remains unclear. In this study, we generated smooth muscle-specific LGL1 knockout (LGL1SMKO) mice by cross-breeding LGL1flox/flox mice with α-SMA-Cre mice. LGL1 level was significantly decreased during calcifying conditions. Overexpression of LGL1 restrained high phosphate-induced calcification in vascular smooth muscle cells (VSMCs). Mechanically, LGL1 could bind with high mobility group box 1 (HMGB1) and promote its degradation via the lysosomal pathway, thereby inhibiting calcification. Smooth muscle-specific deletion of LGL1 increased HMGB1 level and aggravated vitamin D3-induced vascular calcification, which was attenuated by an HMGB1 inhibitor. LGL1 may inhibit vascular calcification by preventing osteogenic differentiation via promoting HMGB1 degradation.


Subject(s)
Calcinosis/etiology , Glycoproteins/genetics , HMGB1 Protein/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers , Calcinosis/metabolism , Cells, Cultured , Disease Models, Animal , Gene Expression , Glycoproteins/deficiency , Glycoproteins/metabolism , HMGB1 Protein/metabolism , Humans , Immunohistochemistry , Mice , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Protein Binding , Vitamin D/metabolism
9.
Mol Vis ; 26: 563-573, 2020.
Article in English | MEDLINE | ID: mdl-32818018

ABSTRACT

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.


Subject(s)
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
10.
Mol Cell ; 48(1): 39-51, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22981861

ABSTRACT

Endoplasmic reticulum (ER) thiol oxidases initiate a disulfide relay to oxidatively fold secreted proteins. We found that combined loss-of-function mutations in genes encoding the ER thiol oxidases ERO1α, ERO1ß, and PRDX4 compromised the extracellular matrix in mice and interfered with the intracellular maturation of procollagen. These severe abnormalities were associated with an unexpectedly modest delay in disulfide bond formation in secreted proteins but a profound, 5-fold lower procollagen 4-hydroxyproline content and enhanced cysteinyl sulfenic acid modification of ER proteins. Tissue ascorbic acid content was lower in mutant mice, and ascorbic acid supplementation improved procollagen maturation and lowered sulfenic acid content in vivo. In vitro, the presence of a sulfenic acid donor accelerated the oxidative inactivation of ascorbate by an H(2)O(2)-generating system. Compromised ER disulfide relay thus exposes protein thiols to competing oxidation to sulfenic acid, resulting in depletion of ascorbic acid, impaired procollagen proline 4-hydroxylation, and a noncanonical form of scurvy.


Subject(s)
Ascorbic Acid/metabolism , Endoplasmic Reticulum/metabolism , Glycoproteins/metabolism , Oxidoreductases/metabolism , Scurvy/etiology , Scurvy/metabolism , Animals , Ascorbic Acid/pharmacology , Cells, Cultured , Connective Tissue/metabolism , Connective Tissue/pathology , Disease Models, Animal , Disulfides/metabolism , Female , Glycoproteins/deficiency , Glycoproteins/genetics , Male , Mice , Mice, Mutant Strains , Mutation , Oxidation-Reduction , Oxidoreductases/deficiency , Oxidoreductases/genetics , Peroxiredoxins/deficiency , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Procollagen/metabolism , Protein Folding , Protein Processing, Post-Translational/drug effects , Scurvy/genetics , Scurvy/pathology , Sulfenic Acids/metabolism , Transforming Growth Factor beta/metabolism
11.
J Immunol ; 198(5): 1805-1814, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28108561

ABSTRACT

Semaphorin 3E (Sema3E) plays a crucial role in axon guidance, vascular patterning, and immune regulation. Nevertheless, the role of Sema3E in asthma is still elusive. In this study, we show that genetic ablation of Sema3E in mice results in increased lung granulocytosis, airway hyperresponsiveness, mucus overproduction, collagen deposition, and Th2/Th17 inflammation. Transfer of Sema3e-/- bone marrow progenitor cells to irradiated wild-type (WT) recipients exacerbates airway hyperresponsiveness and inflammation, whereas transfer of WT bone marrow progenitor cells ameliorates asthma pathology in Sema3e-/- recipients. Sema3e-/- mice display a higher frequency of CD11b+ pulmonary dendritic cells than their WT controls at the baseline and after sensitization with house dust mite. Adoptive transfer of CD11b+ pulmonary dendritic cells from Sema3e-/- mice into WT recipients increases house dust mite-induced Th2/Th17 inflammation in the airway. Together, these findings identify Sema3E as a novel regulatory molecule in allergic asthma that acts upstream of proallergic events and suggest that targeting this molecule could be a novel approach to treat allergic asthma.


Subject(s)
Allergens/immunology , Asthma/immunology , Glycoproteins/deficiency , Glycoproteins/physiology , Inflammation/immunology , Membrane Proteins/deficiency , Membrane Proteins/physiology , Respiratory Hypersensitivity/immunology , Adoptive Transfer , Animals , Cytokines/biosynthesis , Cytokines/immunology , Cytoskeletal Proteins , Dendritic Cells/immunology , Disease Models, Animal , Gene Expression Regulation , Glycoproteins/genetics , Lung/immunology , Lung/physiopathology , Membrane Proteins/genetics , Mice , Pyroglyphidae/immunology , Semaphorins , Th17 Cells/immunology , Th17 Cells/metabolism
12.
J Cell Mol Med ; 22(1): 684-694, 2018 01.
Article in English | MEDLINE | ID: mdl-28990324

ABSTRACT

Stanniocalcins are expressed in the pancreas tissue, and it was suggested a direct correlation between circulating insulin and STC2 concentrations in human. Here, we show a significant correlation between STC1 and both glycaemia and glycosylated haemoglobin among DM2 patients, while DM2 patients who present the greatest glycosylated haemoglobin values exhibited the lowest STC2 expression. However, treatment of patients with antiglycaemic drugs does not significantly modify the expression of both STCs. On the other hand, STC2-/- mice that exhibited neonatal and adult overweight further presented deregulated glycaemia when they were feed with a hypercaloric diet (breeding pellet, BP). This alteration is more evident at the early stages of the animal life. Deregulated glycaemia in these mice was confirmed using glucose oral test. In addition, STC2-/- mice present enhanced pancreas size; thus, the histological analysis reveals that WT mice respond to BP diet by increasing the size of the pancreatic islets through inducing cell division, and STC2-/- mice lack this compensatory mechanism. Contrary, BP fed STC2-/- mice show enhanced number of islets but of similar size than those fed with regular pellet. Histopathological analysis demonstrates tissue structure disruption and erythrocytes infiltrations in STC2-/- mice, possibly due to the stress evoked by the BP diet. Finally, enhanced glucagon immunostaining was observed in the islet of STC2-/- mice, and the glucagon ELISA assay confirmed the increase in the circulating glucagon. Summarizing, we present evidence of the role of STCs, mainly STC2, as a possible early marker during development of diabetes mellitus.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Adult , Aged , Animals , Glucagon/blood , Glycoproteins/deficiency , Humans , Intracellular Signaling Peptides and Proteins , Mice, Inbred C57BL , Mice, Obese , Middle Aged , Organ Size , Pancreas/metabolism , Pancreas/pathology
13.
Dev Biol ; 426(2): 245-254, 2017 06 15.
Article in English | MEDLINE | ID: mdl-27364468

ABSTRACT

The dorsal ventral axis of vertebrates requires high BMP activity for ventral development and inhibition of BMP activity for dorsal development. Presumptive dorsal regions of the embryo are protected from the ventralizing activity of BMPs by the secretion of BMP antagonists from the mesoderm. Noggin, one such antagonist, binds BMP ligands and prevents them from binding their receptors, however, a unique role for Noggin in amphibian development has remained unclear. Previously, we used zinc-finger nucleases to mutagenize the noggin locus in Xenopus tropicalis. Here, we report on the phenotype of noggin mutant frogs as a result of breeding null mutations to homozygosity. Early homozygous noggin mutant embryos are indistinguishable from wildtype siblings, with normal neural induction and neural tube closure. However, in late tadpole stages mutants present severe ventral craniofacial defects, notably a fusion of Meckel's cartilage to the palatoquadrate cartilage. Consistent with a noggin loss-of-function, mutants show expansions of BMP target gene expression and the mutant phenotype can be rescued with transient BMP inhibition. These results demonstrate that in amphibians, Noggin is dispensable for early embryonic patterning but is critical for cranial skeletogenesis.


Subject(s)
Branchial Region/growth & development , Carrier Proteins/physiology , Xenopus Proteins/physiology , Xenopus/growth & development , Alleles , Animals , Body Patterning , Bone Morphogenetic Proteins/pharmacology , Bone Morphogenetic Proteins/physiology , Carrier Proteins/genetics , Cartilage/abnormalities , Cell Differentiation , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Follistatin/deficiency , Follistatin/genetics , Gene Knockout Techniques , Glycoproteins/deficiency , Glycoproteins/genetics , Homozygote , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Larva , Mandible/abnormalities , Morpholinos/pharmacology , Skull/abnormalities , Xenopus/embryology , Xenopus Proteins/deficiency , Xenopus Proteins/genetics
14.
J Cell Physiol ; 233(2): 1156-1167, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28460416

ABSTRACT

The bone marrow niche is a dynamic and complex microenvironment that can both regulate, and be regulated by the bone matrix. Within the bone marrow (BM), mesenchymal stromal cell (MSC) precursors reside in a multi-potent state and retain the capacity to differentiate down osteoblastic, adipogenic, or chondrogenic lineages in response to numerous biochemical cues. These signals can be altered in various pathological states including, but not limited to, osteoporotic-induced fracture, systemic adiposity, and the presence of bone-homing cancers. Herein we provide evidence that signals from the bone matrix (osteocytes) determine marrow adiposity by regulating adipogenesis in the bone marrow. Specifically, we found that physiologically relevant levels of Sclerostin (SOST), which is a Wnt-inhibitory molecule secreted from bone matrix-embedded osteocytes, can induce adipogenesis in 3T3-L1 cells, mouse ear- and BM-derived MSCs, and human BM-derived MSCs. We demonstrate that the mechanism of SOST induction of adipogenesis is through inhibition of Wnt signaling in pre-adipocytes. We also demonstrate that a decrease of sclerostin in vivo, via both genetic and pharmaceutical methods, significantly decreases bone marrow adipose tissue (BMAT) formation. Overall, this work demonstrates a direct role for SOST in regulating fate determination of BM-adipocyte progenitors. This provides a novel mechanism for which BMAT is governed by the local bone microenvironment, which may prove relevant in the pathogenesis of certain diseases involving marrow adipose. Importantly, with anti-sclerostin therapy at the forefront of osteoporosis treatment and a greater recognition of the role of BMAT in disease, these data are likely to have important clinical implications.


Subject(s)
Adipocytes/metabolism , Adipogenesis , Adipose Tissue/metabolism , Bone Marrow Cells/metabolism , Glycoproteins/metabolism , Mesenchymal Stem Cells/metabolism , Osteocytes/metabolism , 3T3-L1 Cells , Adaptor Proteins, Signal Transducing , Adipose Tissue/cytology , Adiposity , Animals , Culture Media, Conditioned/metabolism , Glycoproteins/deficiency , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Paracrine Communication , Phenotype , Stem Cell Niche , Wnt Signaling Pathway
15.
Circulation ; 136(22): 2162-2177, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-28978553

ABSTRACT

BACKGROUND: Cell therapy remains the most promising approach against ischemic heart injury. However, the poor survival of engrafted stem cells in the ischemic environment limits their therapeutic efficacy for cardiac repair after myocardial infarction. CTRP9 (C1q/tumor necrosis factor-related protein-9) is a novel prosurvival cardiokine with significantly downregulated expression after myocardial infarction. Here we tested a hypothesis that CTRP9 might be a cardiokine required for a healthy microenvironment promoting implanted stem cell survival and cardioprotection. METHODS: Mice were subjected to myocardial infarction and treated with adipose-derived mesenchymal stem cells (ADSCs, intramyocardial transplantation), CTRP9, or their combination. Survival, cardiac remodeling and function, cardiomyocytes apoptosis, and ADSCs engraftment were evaluated. Whether CTRP9 directly regulates ADSCs function was determined in vitro. Discovery-drive approaches followed by cause-effect analysis were used to uncover the molecular mechanisms of CTRP9. RESULTS: Administration of ADSCs alone failed to exert significant cardioprotection. However, administration of ADSCs in addition to CTRP9 further enhanced the cardioprotective effect of CTRP9 (P<0.05 or P<0.01 versus CTRP9 alone), suggesting a synergistic effect. Administration of CTRP9 at a dose recovering physiological CTRP9 levels significantly prolonged ADSCs retention/survival after implantation. Conversely, the number of engrafted ADSCs was significantly reduced in the CTRP9 knockout heart. In vitro study demonstrated that CTRP9 promoted ADSCs proliferation and migration, and it protected ADSCs against hydrogen peroxide-induced cellular death. CTRP9 enhances ADSCs proliferation/migration by extracellular regulated protein kinases (ERK)1/2-matrix metallopeptidase 9 signaling and promotes antiapoptotic/cell survival via ERK-nuclear factor erythroid-derived 2-like 2/antioxidative protein expression. N-cadherin was identified as a novel CTRP9 receptor mediating ADSCs signaling. Blockade of either N-cadherin or ERK1/2 completely abolished the previously noted CTRP9 effects. Although CTRP9 failed to promote ADSCs cardiogenic differentiation, CTRP9 promotes superoxide dismutase 3 expression and secretion from ADSCs, protecting cardiomyocytes against oxidative stress-induced cell death. CONCLUSIONS: We provide the first evidence that CTRP9 promotes ADSCs proliferation/survival, stimulates ADSCs migration, and attenuates cardiomyocyte cell death by previously unrecognized signaling mechanisms. These include binding with N-cadherin, activation of ERK-matrix metallopeptidase 9 and ERK-nuclear factor erythroid-derived 2-like 2 signaling, and upregulation/secretion of antioxidative proteins. These results suggest that CTRP9 is a cardiokine critical in maintaining a healthy microenvironment facilitating stem cell engraftment in infarcted myocardial tissue, thereby enhancing stem cell therapeutic efficacy.


Subject(s)
Adiponectin/metabolism , Glycoproteins/metabolism , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/prevention & control , Myocytes, Cardiac/metabolism , Regeneration , Signal Transduction , Adiponectin/administration & dosage , Adiponectin/deficiency , Adiponectin/genetics , Adipose Tissue/cytology , Animals , Apoptosis , Cadherins/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Glycoproteins/administration & dosage , Glycoproteins/deficiency , Glycoproteins/genetics , Green Fluorescent Proteins/genetics , Hydrogen Peroxide/toxicity , Male , Matrix Metalloproteinase 9/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NF-E2-Related Factor 2/metabolism , Phenotype , Regeneration/drug effects , Signal Transduction/drug effects , Stem Cell Niche , Superoxide Dismutase/metabolism , Time Factors
16.
Cell Immunol ; 331: 9-15, 2018 09.
Article in English | MEDLINE | ID: mdl-29748000

ABSTRACT

The present study was conducted to characterize the C6orf120 gene, by using C6orf120 gene-deleted rats (C6orf120-/-), to determine its role in the development and severity of autoimmune hepatitis induced by concanavalin A (Con A), as well as the underlying mechanisms. We found that following Con A injection, C6orf120-/- rats were less susceptible to developing autoimmune hepatitis with low levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) post challenge. Additionally, C6orf120 deficiency increased the frequency of cluster of differentiation (CD)4+ CD25+ Forkhead box P3+ regulatory T cells (Tregs) among intrahepatic lymphocytes, splenocytes, peripheral blood mononuclear cells, and CD4+ T in vitro. Moreover, C6orf120 deficiency downregulated interleukin (IL)-1ß, IL-6, tumor necrosis factor alpha-α, interferon-γ and IL-17a secretion in the plasma and liver tissues. Our results indicated that the C6orf120 gene-deleted rats were less susceptible to Con A-induced autoimmune hepatitis, which may be partly related to the increased frequency of Tregs and inhibited secretion of inflammatory cytokines.


Subject(s)
Gene Deletion , Glycoproteins/genetics , Hepatitis, Autoimmune/genetics , T-Lymphocytes, Regulatory/metabolism , Animals , Concanavalin A/toxicity , Cytokines/metabolism , Glycoproteins/deficiency , Hepatitis, Autoimmune/etiology , Hepatitis, Autoimmune/metabolism , Inflammation Mediators/metabolism , Leukocytes, Mononuclear/metabolism , Liver/metabolism , Liver/pathology , Male , Rats, Sprague-Dawley
17.
Acta Neurol Scand ; 137(3): 308-315, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29143313

ABSTRACT

OBJECTIVES: Disorders of glycogen metabolism include rare hereditary muscle glycogen storage diseases with polyglucosan, which are characterized by storage of abnormally structured glycogen in muscle in addition to exercise intolerance or muscle weakness. In this study, we investigated the etiology and pathogenesis of a late-onset myopathy associated with glycogenin-1 deficiency. MATERIALS AND METHODS: A family with two affected siblings, 64- and 66-year-olds, was studied. Clinical examination and whole-body MRI revealed weakness and wasting in the hip girdle and proximal leg muscles affecting ambulation in the brother. The sister had weakness and atrophy of hands and slight foot dorsiflexion difficulties. Muscle biopsy and whole-exome sequencing were performed in both cases to identify and characterize the pathogenesis including the functional effects of identified mutations. RESULTS: Both siblings demonstrated storage of glycogen that was partly resistant to alpha-amylase digestion. Both were heterozygous for two mutations in GYG1, one truncating 1-base deletion (c.484delG; p.Asp163Thrfs*5) and one novel missense mutation (c.403G>A; p.Gly135Arg). The mutations caused reduced expression of glycogenin-1 protein, and the missense mutation abolished the enzymatic function as analyzed by an in vitro autoglucosylation assay. CONCLUSION: We present functional evidence for the pathogenicity of a novel GYG1 missense mutation located in the substrate binding domain. Our results also demonstrate that glycogenin-1 deficiency may present with highly variable distribution of weakness and wasting also in the same family.


Subject(s)
Glucans/metabolism , Glucosyltransferases/genetics , Glycogen Storage Disease/genetics , Glycoproteins/genetics , Muscular Diseases/genetics , Aged , Female , Glucosyltransferases/deficiency , Glycogen Storage Disease/pathology , Glycoproteins/deficiency , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle Weakness/genetics , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Mutation, Missense , Pedigree , Siblings
18.
Clin Endocrinol (Oxf) ; 87(3): 300-311, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28445628

ABSTRACT

OBJECTIVE: Acid-labile subunit deficiency (ACLSD), caused by inactivating mutations in both IGFALS gene alleles, is characterized by marked reduction in IGF-I and IGFBP-3 levels associated with mild growth retardation. The aim of this study was to expand the known phenotype and genetic characteristics of ACLSD by reporting data from four index cases and their families. DESIGN: Auxological data, biochemical and genetic studies were performed in four children diagnosed with ACLSD and all available relatives. METHODS: Serum levels of IGF-I, IGFBP-3, acid-labile subunit (ALS), and in vitro ternary complex formation (ivTCF) were determined. After sequencing the IGFALS gene, pathogenicity of novel identified variants was evaluated by in vitro expression in transfected Chinese hamster ovarian (CHO) cells. ALS protein was detected in patients' sera and CHO cells conditioned media and lysates by Western immunoblot (WIB). RESULTS: Four index cases and four relatives were diagnosed with ACLSD. The following variants were found: p.Glu35Glyfs*17, p.Glu35Lysfs*87, p.Leu213Phe, p.Asn276Ser, p.Leu409Phe, p.Ala475Val and p.Ser490Trp. ACLSD patients presented low IGF-I and low or undetectable levels of IGFBP-3 and ALS. Seven out of 8 patients did not form ivTCF. CONCLUSIONS: This study confirms previous findings in ACLSD, such as the low IGF-I and a more severe reduction in IGFBP-3 levels, and a gene dosage effect observed in heterozygous carriers (HC). In addition, father-to-son transmission (father compound heterozygous and mother HC), preservation of male fertility, and marginal ALS expression with potential involvement in preserved responsiveness to rhGH treatment, are all novel aspects, not previously reported in this condition.


Subject(s)
Glycoproteins/deficiency , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/analysis , Adolescent , Adult , Aged , Animals , Carrier Proteins/genetics , Child , Child, Preschool , Cricetulus , Family , Female , Fertility , Genetic Variation , Glycoproteins/genetics , Growth Disorders/genetics , Heterozygote , Humans , Infant , Insulin-Like Growth Factor Binding Protein 3/deficiency , Insulin-Like Growth Factor I/deficiency , Latin America , Male , Middle Aged , Mutation , Transfection , Young Adult
19.
J Inherit Metab Dis ; 40(1): 139-149, 2017 01.
Article in English | MEDLINE | ID: mdl-27718144

ABSTRACT

We describe a new type of cardiomyopathy caused by a mutation in the glycogenin-1 gene (GYG1). Three unrelated male patients aged 34 to 52 years with cardiomyopathy and abnormal glycogen storage on endomyocardial biopsy were homozygous for the missense mutation p.Asp102His in GYG1. The mutated glycogenin-1 protein was expressed in cardiac tissue but had lost its ability to autoglucosylate as demonstrated by an in vitro assay and western blot analysis. It was therefore unable to form the primer for normal glycogen synthesis. Two of the patients showed similar patterns of heart dilatation, reduced ejection fraction and extensive late gadolinium enhancement on cardiac magnetic resonance imaging. These two patients were severely affected, necessitating cardiac transplantation. The cardiomyocyte storage material was characterized by large inclusions of periodic acid and Schiff positive material that was partly resistant to alpha-amylase treatment consistent with polyglucosan. The storage material had, unlike normal glycogen, a partly fibrillar structure by electron microscopy. None of the patients showed signs or symptoms of muscle weakness but a skeletal muscle biopsy in one case revealed muscle fibres with abnormal glycogen storage. Glycogenin-1 deficiency is known as a rare cause of skeletal muscle glycogen storage disease, usually without cardiomyopathy. We demonstrate that it may also be the cause of severe cardiomyopathy and cardiac failure without skeletal muscle weakness. GYG1 should be included in cardiomyopathy gene panels.


Subject(s)
Cardiomyopathies/genetics , Glucosyltransferases/deficiency , Glucosyltransferases/genetics , Glycoproteins/deficiency , Glycoproteins/genetics , Mutation, Missense/genetics , Adult , Biopsy , Glucans/genetics , Glycogen/genetics , Glycogen Storage Disease/genetics , Homozygote , Humans , Male , Middle Aged , Muscle, Skeletal/metabolism
20.
Arterioscler Thromb Vasc Biol ; 36(1): 122-33, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26564819

ABSTRACT

OBJECTIVE: Arterial injury stimulates remodeling responses that, when excessive, lead to stenosis. These responses are influenced by integrin signaling in vascular smooth muscle cells (VSMCs). Microfibrillar-associated protein 4 (MFAP4) is an integrin ligand localized to extracellular matrix fibers in the vascular wall. The role of MFAP4 in vascular biology is unknown. We aimed to test the hypothesis that MFAP4 would enhance integrin-dependent VSMC activation. APPROACH AND RESULTS: We produced Mfap4-deficient (Mfap4(-/-)) mice and performed carotid artery ligation to explore the role of MFAP4 in vascular biology in vivo. Furthermore, we investigated the effects of MFAP4 in neointimal formation ex vivo and in primary VSMC and monocyte cultures in vitro. When challenged with carotid artery ligation, Mfap4(-/-) mice exhibited delayed neointimal formation, accompanied by early reduction in the number of proliferating medial and neointimal cells, as well as infiltrating leukocytes. Delayed neointimal formation was associated with decreased cross-sectional area of ligated Mfap4(-/-) carotid arteries resulting in lumen narrowing 28 days after ligation. MFAP4 blockade prohibited the formation of neointimal hyperplasia ex vivo. Moreover, we demonstrated that MFAP4 is a ligand for integrin αVß3 and mediates VSMC phosphorylation of focal adhesion kinase, migration, and proliferation in vitro. MFAP4-dependent VSMC activation was reversible by treatment with MFAP4-blocking antibodies and inhibitors of focal adhesion kinase and downstream kinases. In addition, we showed that MFAP4 promotes monocyte chemotaxis in integrin αVß3-dependent manner. CONCLUSIONS: MFAP4 regulates integrin αVß3-induced VSMC proliferation and migration, as well as monocyte chemotaxis, and accelerates neointimal hyperplasia after vascular injury.


Subject(s)
Carotid Artery Diseases/metabolism , Carrier Proteins/metabolism , Cell Movement , Cell Proliferation , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Animals , Apoptosis , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Carrier Proteins/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemotaxis, Leukocyte , Disease Models, Animal , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/metabolism , Genotype , Glycoproteins/deficiency , Glycoproteins/genetics , Humans , Hyperplasia , Integrin alphaVbeta3/metabolism , Ligands , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Phenotype , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Time Factors , Vascular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL