Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
J Reprod Dev ; 70(2): 55-64, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38246612

ABSTRACT

The mammalian X chromosome exhibits enrichment in genes associated with germ cell development. Previously, we generated a rat model of Becker muscular dystrophy (BMD) characterized by an in-frame mutation in the dystrophin gene, situated on the X chromosome and responsible for encoding a protein crucial for muscle integrity. Male BMD rats are infertile owing to the absence of normal spermatids in the epididymis. Within the seminiferous tubules of BMD rats, elongated spermatids displayed abnormal morphology. To elucidate the cause of infertility, we identified a putative gene containing an open reading frame situated in the intronic region between exons 6 and 7 of the dystrophin gene, specifically deleted in male BMD rats. This identified gene, along with its encoded protein, exhibited specific detection within the testes, exclusively localized in round to elongated spermatids during spermiogenesis. Consequently, we designated the encoded protein as dystrophin-locus-derived testis-specific protein (DTSP). Given the absence of DTSP in the testes of BMD rats, we hypothesized that the loss of DTSP contributes to the infertility observed in male BMD rats.


Subject(s)
Infertility , Succinimides , Testis , Male , Rats , Animals , Testis/metabolism , Dystrophin/genetics , Dystrophin/metabolism , Spermatogenesis/genetics , Proteins/metabolism , Infertility/metabolism , Mammals
2.
Mol Cell Endocrinol ; 576: 112008, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37422125

ABSTRACT

We established a methodology using machine learning algorithms for determining the pathogenic factors for premenstrual dysphoric disorder (PMDD). PMDD is a disease characterized by emotional and physical symptoms that occurs before menstruation in women of childbearing age. Owing to the diverse manifestations and various pathogenic factors associated with this disease, the diagnosis of PMDD is time-consuming and challenging. In the present study, we aimed to establish a methodology for diagnosing PMDD. Using an unsupervised machine-learning algorithm, we divided pseudopregnant rats into three clusters (C1 to C3), depending on the level of anxiety- and depression-like behaviors. From the results of RNA-seq and subsequent qPCR of the hippocampus in each cluster, we identified 17 key genes for building a PMDD diagnostic model using our original two-step feature selection with supervised machine learning. By inputting the expression levels of these 17 genes into the machine learning classifier, the PMDD symptoms of another group of rats were successfully classified as C1-C3 with an accuracy of 96%, corresponding to the classification by behavior. The present methodology would be applicable for the clinical diagnosis of PMDD using blood samples instead of samples from the hippocampus in the future.


Subject(s)
Premenstrual Dysphoric Disorder , Premenstrual Syndrome , Humans , Female , Animals , Rats , Premenstrual Dysphoric Disorder/diagnosis , Premenstrual Dysphoric Disorder/metabolism , Premenstrual Dysphoric Disorder/psychology , Premenstrual Syndrome/diagnosis , Premenstrual Syndrome/psychology , Emotions , Machine Learning , Algorithms
3.
Methods Mol Biol ; 2631: 325-340, 2023.
Article in English | MEDLINE | ID: mdl-36995675

ABSTRACT

The clustered regularly interspaced short palindromic repeats (CRISPR) technology has made it possible to produce genome-edited (GE) animals more easily and rapidly than before. In most cases, GE mice are produced by microinjection (MI) or by in vitro electroporation (EP) of CRISPR reagents into fertilized eggs (zygotes). Both of these approaches require ex vivo handling of isolated embryos and their subsequent transfer into another set of mice (called recipient or pseudopregnant mice). Such experiments are performed by highly skilled technicians (especially for MI). We recently developed a novel genome editing method, called "GONAD (Genome-editing via Oviductal Nucleic Acids Delivery)," which can completely eliminate the ex vivo handling of embryos. We also made improvements to the GONAD method, termed "improved-GONAD (i-GONAD)." The i-GONAD method involves injection of CRISPR reagents into the oviduct of an anesthetized pregnant female using a mouthpiece-controlled glass micropipette under a dissecting microscope, followed by EP of the entire oviduct allowing the CRISPR reagents to enter into the zygotes present inside the oviduct, in situ. After the i-GONAD procedure, the mouse recovered from anesthesia is allowed to continue the pregnancy to full term to deliver its pups. The i-GONAD method does not require pseudopregnant female animals for embryo transfer, unlike the methods relying on ex vivo handling of zygotes. Therefore, the i-GONAD method can reduce the number of animals used, compared to the traditional methods. In this chapter, we describe some newer technical tips about the i-GONAD method. Additionally, even though the detailed protocols of GONAD and i-GONAD have been published elsewhere (Gurumurthy et al., Curr Protoc Hum Genet 88:15.8.1-15.8.12, 2016 Nat Protoc 14:2452-2482, 2019), we provide all the protocol steps of i-GONAD in this chapter so that the reader can find most of the information, needed for performing i-GONAD experiments, in one place.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Humans , Pregnancy , Female , Mice , Animals , Gene Editing/methods , CRISPR-Cas Systems/genetics , Fallopian Tubes , Oviducts , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Electroporation/methods , Gonads
4.
Skelet Muscle ; 12(1): 24, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36258243

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is an X-linked muscle disease caused by a complete lack of dystrophin, which stabilizes the plasma membrane of myofibers. The orofacial function is affected in an advanced stage of DMD and this often leads to an eating disorder such as dysphagia. Dysphagia is caused by multiple etiologies including decreased mastication and swallowing. Therefore, preventing the functional declines of mastication and swallowing in DMD is important to improve the patient's quality of life. In the present study, using a rat model of DMD we generated previously, we performed analyses on the masseter and tongue muscles, both are required for proper eating function. METHODS: Age-related changes of the masseter and tongue muscle of DMD rats were analyzed morphometrically, histologically, and immunohistochemically. Also, transcription of cellular senescent markers, and utrophin (Utrn), a functional analog of dystrophin, was examined. RESULTS: The masseter muscle of DMD rats showed progressive dystrophic changes as observed in their hindlimb muscle, accompanied by increased transcription of p16 and p19. On the other hand, the tongue of DMD rats showed macroglossia due to hypertrophy of myofibers with less dystrophic changes. Proliferative activity was preserved in the satellite cells from the tongue muscle but was perturbed severely in those from the masseter muscle. While Utrn transcription was increased in the masseter muscle of DMD rats compared to WT rats, probably due to a compensatory mechanism, its level in the tongue muscle was comparable between WT and DMD rats and was similar to that in the masseter muscle of DMD rats. CONCLUSIONS: Muscular dystrophy is less advanced in the tongue muscle compared to the masseter muscle in the DMD rat.


Subject(s)
Deglutition Disorders , Macroglossia , Muscular Dystrophy, Duchenne , Mice , Rats , Animals , Dystrophin/genetics , Dystrophin/metabolism , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Utrophin/metabolism , Mice, Inbred mdx , Macroglossia/etiology , Macroglossia/pathology , Deglutition Disorders/metabolism , Deglutition Disorders/pathology , Quality of Life , Muscle, Skeletal/metabolism , Tongue
5.
J Vet Med Sci ; 83(9): 1416-1424, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34334511

ABSTRACT

Rhabdomyosarcoma (RMS) is an aggressive type of soft tissue sarcoma, and pleomorphic RMS is a rare subtype of RMS found in adult. p16 is a tumor suppressor which inhibits cell cycle. In human RMS, p16 gene is frequently deleted, but p16-null mice do not develop RMS. We reported that genetic ablation of p16 by the crossbreeding of p16 knock-out rats (p16-KO rats) improved the dystrophic phenotype of a rat model of Duchenne muscular dystrophy (Dmd-KO rats). However, p16/Dmd double knock-out rats (dKO rats) unexpectedly developed sarcoma. In the present study, we raised p16-KO, Dmd-KO, and dKO rats until 11 months of age. Twelve out of 22 dKO rats developed pleomorphic RMS after 9 months of age, while none of p16-KO rats and Dmd-KO rats developed tumor. The neoplasms were connected to skeletal muscle tissue with indistinct borders and characterized by diffuse proliferation of pleomorphic cells which had eosinophilic cytoplasm and atypical nuclei with anisokaryosis. For almost all cases, the tumor cells immunohistochemically expressed myogenic markers including desmin, MyoD, and myogenin. The single cell cloning from tumor primary cells gained 20 individual Pax7-negative MyoD-positive RMS cell clones. Our results demonstrated that double knock-out of p16 and dystrophin in rats leads to the development of pleomorphic RMS, providing an animal model that may be useful to study the developmental mechanism of pleomorphic RMS.


Subject(s)
Muscular Dystrophy, Duchenne , Rhabdomyosarcoma , Rodent Diseases , Sarcoma , Animals , Cyclin-Dependent Kinase Inhibitor p16 , Dystrophin/genetics , Mice , Muscle, Skeletal , Rats , Rhabdomyosarcoma/genetics
6.
Anim Sci J ; 92(1): e13573, 2021.
Article in English | MEDLINE | ID: mdl-34231933

ABSTRACT

The degree of intramuscular adipose tissue accumulation is one of the factors affecting meat quality. Accumulation of adipocytes is also observed under the pathological condition of skeletal muscle such as muscular dystrophy and sarcopenia. The origin of adipocytes seen in skeletal muscle is mesenchymal progenitor cells that can give rise to both adipocytes and fibroblasts. In the present study, we demonstrated that siRNA-mediated suppression of MyoD expression in rat skeletal muscle progenitor cell culture, which comprises both myogenic satellite cells and mesenchymal progenitor cells, resulted in diminished myotube formation and an unexpected spontaneous appearance of white adipocytes. Suppressing myomaker expression also resulted in complete absence of myotube formation without reducing MyoD expression, but no adipogenesis was seen in this scenario, indicating that decline in MyoD expression rather than decreased myotube formation is necessary to induce adipogenesis. In addition, spontaneous adipogenesis induced by suppressing MyoD expression in culture was inhibited by the conditioned medium from control culture, indicating that anti-adipogenic factor(s) are secreted from MyoD-positive myogenic cells. These results indicate the presence of regulatory mechanism on adipogenesis by myogenic cells.


Subject(s)
Adipogenesis , Satellite Cells, Skeletal Muscle , Adipogenesis/genetics , Animals , Muscle Fibers, Skeletal , Muscle, Skeletal , Rats , Stem Cells
7.
Sci Rep ; 10(1): 16385, 2020 10 12.
Article in English | MEDLINE | ID: mdl-33046751

ABSTRACT

Duchenne muscular dystrophy (DMD) is a progressive disease characterised by chronic muscle degeneration and inflammation. Our previously established DMD model rats (DMD rats) have a more severe disease phenotype than the broadly used mouse model. We aimed to investigate the role of senescence in DMD using DMD rats and patients. Senescence was induced in satellite cells and mesenchymal progenitor cells, owing to the increased expression of CDKN2A, p16- and p19-encoding gene. Genetic ablation of p16 in DMD rats dramatically restored body weight and muscle strength. Histological analysis showed a reduction of fibrotic and adipose tissues invading skeletal muscle, with increased muscle regeneration. Senolytic drug ABT263 prevented loss of body weight and muscle strength, and increased muscle regeneration in rats even at 8 months-the late stage of DMD. Moreover, senescence markers were highly expressed in the skeletal muscle of DMD patients. In situ hybridization of CDKN2A confirmed the expression of it in satellite cells and mesenchymal progenitor cells in patients with DMD. Collectively, these data provide new insights into the integral role of senescence in DMD progression.


Subject(s)
Cellular Senescence/genetics , Disease Models, Animal , Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Mutation , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Dystrophin/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/metabolism , Rats , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism
8.
Dis Model Mech ; 13(9)2020 09 28.
Article in English | MEDLINE | ID: mdl-32859695

ABSTRACT

Dystrophin, encoded by the DMD gene on the X chromosome, stabilizes the sarcolemma by linking the actin cytoskeleton with the dystrophin-glycoprotein complex (DGC). In-frame mutations in DMD cause a milder form of X-linked muscular dystrophy, called Becker muscular dystrophy (BMD), characterized by the reduced expression of truncated dystrophin. So far, no animal model with in-frame mutations in Dmd has been established. As a result, the effect of in-frame mutations on the dystrophin expression profile and disease progression of BMD remains unclear. In this study, we established a novel rat model carrying in-frame Dmd gene mutations (IF rats) and evaluated the pathology. We found that IF rats exhibited reduced expression of truncated dystrophin in a proteasome-independent manner. This abnormal dystrophin expression caused dystrophic changes in muscle tissues but did not lead to functional deficiency. We also found that the expression of additional dystrophin named dpX, which forms the DGC in the sarcolemma, was associated with the appearance of truncated dystrophin. In conclusion, the outcomes of this study contribute to the further understanding of BMD pathology and help elucidate the efficiency of dystrophin recovery treatments in Duchenne muscular dystrophy, a more severe form of X-linked muscular dystrophy.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Mutation/genetics , Open Reading Frames/genetics , Animals , Base Sequence , Cell Membrane/metabolism , Disease Models, Animal , Dystroglycans/metabolism , Muscle, Skeletal/pathology , Myocardium/pathology , Phenotype , Protein Isoforms/metabolism , Rats , Sarcolemma/metabolism
9.
Anim Sci J ; 91(1): e13368, 2020.
Article in English | MEDLINE | ID: mdl-32285501

ABSTRACT

Accumulation of intramuscular adipose tissue (IMAT) and development of fibrous tissues due to accumulation of collagen both affect meat quality such as tenderness, texture, and flavor. Thus, it is important for the production of high-quality meat to regulate the amount of adipose and fibrous tissues in skeletal muscle. IMAT is comprised of adipocytes, while collagens included in fibrous tissues are mainly produced by activated fibroblasts. Both adipocytes and fibroblasts are differentiated from their common ancestors, called mesenchymal progenitor cells (MPC). We previously established rat MPC clone, 2G11 cells. As several reports implicated the plasticity of fibroblast differentiation, in the present study, using 2G11 cells, we asked whether myofibroblasts differentiated from MPC are capable of re-gaining adipogenic potential in vitro. By treating with bFGF, their αSMA expression was reduced and adipogenic potential was restored partially. Furthermore, by lowering cell density together with bFGF treatment, 2G11 cell-derived myofibroblasts lost αSMA expression and showed the highest adipogenic potential, and this was along with their morphological change from flattened- to spindle-like shape, which is typically observed with MPC. These results indicated that MPC-derived myofibroblasts could re-acquire adipogenic potential, possibly mediated through returning to an undifferentiated MPC-like state.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/physiology , Muscle, Skeletal/cytology , Myofibroblasts , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Cells, Cultured , Collagen/metabolism , Muscle Development/physiology , Muscle, Skeletal/metabolism , Myofibroblasts/metabolism
10.
Neuroreport ; 31(6): 478-483, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32168097

ABSTRACT

Progranulin (PGRN) is a glycoprotein that is widely expressed among organs, including the central nervous system. PGRN insufficiency is involved in various neurodegenerative disorders such as frontotemporal dementia, Alzheimer's disease, and neuronal ceroid lipofuscinosis. One of the major causes of neuronal damage is hyperactivation of the cerebrum triggered by upregulation of excitatory systems. In the present study, we examined the possible involvement of PGRN in modulating excitability of the cerebrum using wild type and PGRN-deficient mice. First, we treated wild type and PGRN-deficient mice with seizure-inducible drugs, bicuculline or N-methyl-D-aspartate (NMDA), which provoke hyperexcitement of neurons. PGRN-deficient mice showed higher intensity of seizure and longer duration of convulsive behavior when treated with either bicuculline or NMDA. Next, we quantified the expression of NMDA receptor subunits in the hippocampus and cerebral cortex. The expression level of NR2A subunit protein was significantly higher in the hippocampus of PGRN-deficient mice, while no difference was observed in the cerebral cortex. On the other hand, mRNA levels of NMDA receptor subunits in the hippocampus were comparable or even lower in PGRN-deficient mice. These results suggest that PGRN modulates the excitability of the cerebrum by regulating at least partially the protein level of NMDA receptors in the hippocampus.


Subject(s)
Bicuculline/adverse effects , Convulsants/adverse effects , N-Methylaspartate/adverse effects , Progranulins/metabolism , Seizures/genetics , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Progranulins/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/chemically induced , Seizures/metabolism
11.
J Vet Med Sci ; 81(11): 1649-1654, 2019 Nov 29.
Article in English | MEDLINE | ID: mdl-31582603

ABSTRACT

We have previously shown that secreted protein acidic and rich in cysteine (SPARC) promotes myogenic differentiation of rat skeletal muscle progenitor cells in vitro, and in vivo small interfering RNA (siRNA)-mediated transient suppression of SPARC expression in skeletal muscle of mice causes atrophic changes of myofibers, suggesting that SPARC plays a role in the maintenance of skeletal muscle function. In order to know the effect of long-term deficiency of SPARC on skeletal muscle, we performed phenotypic analyses of skeletal muscle of SPARC-null mice. Age-associated changes of myofiber diameters were comparable between wild type (WT) and SPARC-null mice at all ages examined, indicating that the growth of myofibers is unaffected by the absence of SPARC. On the other hand, accumulation of fibrillar collagen was significantly reduced in SPARC-null mice compared to WT mice after 5 months of age without significant changes of collagen I gene expression. The results obtained in the present study suggest that SPARC plays a role to maintain the stiffness of skeletal muscle by regulating collagen accumulation.


Subject(s)
Fibrillar Collagens/metabolism , Muscle, Skeletal/metabolism , Osteonectin/metabolism , Aging/metabolism , Animals , Gene Expression , Male , Mice , Mice, Knockout , Myofibrils , Osteonectin/genetics
12.
Aging (Albany NY) ; 10(4): 747-763, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29695641

ABSTRACT

Sarcopenia is the age-related loss of skeletal muscle mass and function. Skeletal muscle comprises diverse progenitor cells, including mesenchymal progenitor cells (MPCs), which normally support myogenic cell function but cause a decline in skeletal muscle function after differentiating into fibrous/adipose tissue. Cellular senescence is a form of persistent cell cycle arrest caused by cellular stress, including oxidative stress, and is accompanied by the acquisition of senescence-associated secretory phenotype (SASP). Here, we found γH2AX+ senescent cells appeared in the interstitium in skeletal muscle, corresponding in position to that of MPCs. H2O2 mediated oxidative stress in 2G11 cells, a rat MPC clone previously established in our laboratory, successfully induced senescence, as shown by the upregulation of p21 and SASP factors, including IL-6. The senescent 2G11 cells lost their fibro/adipogenic potential, but, intriguingly, coculture of myoblasts with senescent 2G11 cells abrogated the myotube formation, which coincided with the downregulation of myomaker, a muscle-specific protein involved in myogenic cell fusion; however, forced expression of myomaker could not rescue this abrogation. These results suggest that senescent MPCs in aged rat skeletal muscle lose their fibro/adipogenic potential, but differ completely from undifferentiated progenitor cells in that senescent MPCs suppress myoblast fusion and thereby potentially accelerate sarcopenia.


Subject(s)
Cell Differentiation/physiology , Cellular Senescence/physiology , Mesenchymal Stem Cells/pathology , Myoblasts/cytology , Oxidative Stress/physiology , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/physiology , Aging/metabolism , Aging/pathology , Animals , Fibroblasts/cytology , Fibroblasts/metabolism , Male , Mesenchymal Stem Cells/metabolism , Muscle Development/physiology , Myoblasts/metabolism , Rats , Rats, Wistar , Sarcopenia/metabolism , Sarcopenia/pathology
13.
Cytokine ; 108: 17-23, 2018 08.
Article in English | MEDLINE | ID: mdl-29558694

ABSTRACT

Skeletal muscle is an endocrine organ that secretes several proteins, which are collectively termed myokines. Although many studies suggest that exercise regulates myokine secretion, the underlying mechanisms remain unclear and all the exercise-dependent myokines have not yet been identified. Therefore, in this study, we attempted to identify novel exercise-dependent myokines by using our recently developed in vitro contractile model. Differentiated C2C12 myotubes were cultured with or without electrical pulse stimulation (EPS) for 24 h to induce cell contraction, and the myokines secreted in conditioned medium were analyzed using a cytokine array. Although most myokine secretions were not affected by EPS, the secretion of Chemokine (C-C motif) ligand 5 (CCL5) (regulated on activation, normal T cell expressed and secreted (RANTES)) was significantly reduced by EPS. This was further confirmed by ELISA and quantitative PCR. Contraction-dependent calcium transients and activation of 5'-AMP activating protein kinase (AMPK) appears to be involved in this decrease, as the chelating Ca2+ by EGTA blocked contraction-dependent CCL5 reduction, whereas the pharmacological activation of AMPK significantly reduced it. However, Ccl5 gene expression was increased by AMPK activation, suggesting that AMPK-dependent CCL5 decrease occurred via post-transcriptional regulation. Finally, mouse experiments revealed that voluntary wheel-running exercise reduced serum CCL5 levels and Ccl5 gene expression in the fast-twitch muscles. Overall, our study provides the first evidence of an exercise-reducible myokine, CCL5, in the mouse skeletal muscle. Although further studies are required to understand the precise roles of the skeletal muscle cell contraction-induced decrease in CCL5, this decrease may explain some exercise-dependent physiological changes such as those in immune responses.


Subject(s)
Chemokine CCL5/physiology , Muscle Contraction , Muscle, Skeletal/physiology , Physical Conditioning, Animal , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Chemokine CCL5/genetics , Cytokines/genetics , Cytokines/metabolism , Electric Stimulation , Gene Expression , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Real-Time Polymerase Chain Reaction
14.
J Vet Med Sci ; 80(2): 346-353, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29249750

ABSTRACT

Skeletal muscle has an ability to regenerate in response to injury due to the presence of satellite cells. Injury in skeletal muscle causes infiltration of pro-inflammatory macrophages (M1 macrophages) to remove necrotic myofibers, followed by their differentiation into anti-inflammatory macrophages (M2 macrophages) to terminate the inflammation. Since both M1 and M2 macrophages play important roles, coordinated regulation of their kinetics is important to complete muscle regeneration successfully. Progranulin (PGRN) is a pluripotent growth factor, having a protective role against the inflamed tissue. In the central nervous system, PGRN regulates inflammation by inhibiting the activation of microglia. Here we used muscle injury model of PGRN-knockout (PGRN-KO) mice to elucidate whether it has a role in the kinetics of macrophages during muscle regeneration. We found the prolonged persistence of macrophages at the late phase of regeneration in PGRN-KO mice, and these macrophages were suggested to be M2 macrophages since this was accompanied with an increased CD206 expression. We also observed muscle hypertrophy in PGRN-KO mice at the late stage of muscle regeneration. Since M2 macrophages are known to have a role in maturation of myofibers, this muscle hypertrophy may be due to the presence of increased number of M2 macrophages. Our results suggest that PGRN plays a role in the regulation of kinetics of macrophages for the systemic progress of muscle regeneration.


Subject(s)
Intercellular Signaling Peptides and Proteins/deficiency , Macrophages/physiology , Muscle, Skeletal/physiology , Regeneration , Animals , Cobra Cardiotoxin Proteins/pharmacology , Female , Granulins , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Skeletal/physiology , Progranulins
15.
Biosci Biotechnol Biochem ; 82(1): 97-105, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29235416

ABSTRACT

Accumulating evidence indicates that skeletal muscle secrets proteins referred to as myokines and that exercise contributes to their regulation. In this study, we propose that chemokine (C-X-C motif) ligand 10 (CXCL10) functions as a novel myokine. Initially, we stimulated differentiated C2C12 myotubes with or without electrical pulse stimulation (EPS) to identify novel myokines. Cytokine array analysis revealed that CXCL10 secretion was significantly reduced by EPS, which was further confirmed by enzyme-linked immunosorbent assay and quantitative polymerase chain reaction analysis. Treadmill experiments in mice identified significant reduction of Cxcl10 gene expression in the soleus muscle. Additionally, contraction-dependent p38 MAPK activation appeared to be involved in this reduction. Furthermore, C2C12 conditioned medium obtained after applying EPS could induce survival of MSS31, a vascular endothelial cell model, which was partially attenuated by the addition of recombinant CXCL10. Overall, our findings suggest CXCL10 as a novel exercise-reducible myokine, to control endothelial cell viability.


Subject(s)
Chemokine CXCL10/physiology , Exercise Test , Muscle Fibers, Skeletal/physiology , Angiogenesis Inducing Agents , Animals , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression , MAP Kinase Signaling System , Mice , Muscle Contraction , Physical Conditioning, Animal , Polymerase Chain Reaction
16.
Brain Behav Immun ; 66: 165-176, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28655587

ABSTRACT

Sickness responses to lipopolysaccharide (LPS) were examined in mice with deletion of the interleukin (IL)-1 type 1 receptor (IL-1R1). IL-1R1 knockout (KO) mice displayed intact anorexia and HPA-axis activation to intraperitoneally injected LPS (anorexia: 10 or 120µg/kg; HPA-axis: 120µg/kg), but showed attenuated but not extinguished fever (120µg/kg). Brain PGE2 synthesis was attenuated, but Cox-2 induction remained intact. Neither the tumor necrosis factor-α (TNFα) inhibitor etanercept nor the IL-6 receptor antibody tocilizumab abolished the LPS induced fever in IL-1R1 KO mice. Deletion of IL-1R1 specifically in brain endothelial cells attenuated the LPS induced fever, but only during the late, 3rd phase of fever, whereas deletion of IL-1R1 on neural cells or on peripheral nerves had little or no effect on the febrile response. We conclude that while IL-1 signaling is not critical for LPS induced anorexia or stress hormone release, IL-1R1, expressed on brain endothelial cells, contributes to the febrile response to LPS. However, also in the absence of IL-1R1, LPS evokes a febrile response, although this is attenuated. This remaining fever seems not to be mediated by IL-6 receptors or TNFα, but by some yet unidentified pyrogenic factor.


Subject(s)
Anorexia/metabolism , Fever/metabolism , Illness Behavior , Receptors, Interleukin-1 Type I/metabolism , Adrenocorticotropic Hormone/blood , Animals , Anorexia/chemically induced , Brain/metabolism , Corticosterone/blood , Eating , Endothelial Cells/metabolism , Female , Fever/chemically induced , Hypothalamus/metabolism , Inflammation/blood , Inflammation/complications , Inflammation Mediators/blood , Lipopolysaccharides/administration & dosage , Male , Mice, Knockout , Receptors, Interleukin-1 Type I/genetics
17.
Exp Gerontol ; 95: 1-8, 2017 09.
Article in English | MEDLINE | ID: mdl-28479389

ABSTRACT

It is well established that adult neurogenesis in the hippocampus declines with age. Our previous studies have suggested that progranulin (PGRN) has a facilitative effect on hippocampal neurogenesis. We have also shown that PGRN plays a role in suppressing excessive neuroinflammatory responses in the cortex and thalamus after brain injury and aging, respectively. However, the roles of PGRN in modulating neurogenesis and neuroinflammatory responses in the hippocampus of aged animals are not yet understood. In the present study, we investigated neurogenesis and neuroinflammation-related responses in the hippocampus of young (15-week-old) and old (135-week-old) wild-type and PGRN-deficient male mice. Neurogenesis in the dentate gyrus of the hippocampus markedly declined with age, and there was no significant difference between the genotype. The number of CD68-positive activated microglia and the expression of lysosomal genes in the hippocampus were significantly increased with age, and PGRN deficiency further increased them. The expression of pro-inflammatory genes was also increased with age, and PGRN deficiency significantly enhanced some of them. These results suggest that PGRN deficiency exacerbates neuroinflammatory responses related to activated microglia in aged animals, while PGRN may not counteract the decline of hippocampal neurogenesis with age.


Subject(s)
Aging/metabolism , Encephalitis/metabolism , Hippocampus/metabolism , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neurogenesis , Age Factors , Aging/genetics , Aging/pathology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Encephalitis/genetics , Encephalitis/pathology , Granulins , Hippocampus/pathology , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Lysosomes/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Progranulins
18.
J Neurosci ; 37(19): 5035-5044, 2017 05 10.
Article in English | MEDLINE | ID: mdl-28438967

ABSTRACT

Fever occurs upon binding of prostaglandin E2 (PGE2) to EP3 receptors in the median preoptic nucleus of the hypothalamus, but the origin of the pyrogenic PGE2 has not been clearly determined. Here, using mice of both sexes, we examined the role of local versus generalized PGE2 production in the brain for the febrile response. In wild-type mice and in mice with genetic deletion of the prostaglandin synthesizing enzyme cyclooxygenase-2 in the brain endothelium, generated with an inducible CreERT2 under the Slco1c1 promoter, PGE2 levels in the CSF were only weakly related to the magnitude of the febrile response, whereas the PGE2 synthesizing capacity in the hypothalamus, as reflected in the levels of cyclooxygenase-2 mRNA, showed strong correlation with the immune-induced fever. Histological analysis showed that the deletion of cyclooxygenase-2 in brain endothelial cells occurred preferentially in small- and medium-sized vessels deep in the brain parenchyma, such as in the hypothalamus, whereas larger vessels, and particularly those close to the neocortical surface and in the meninges, were left unaffected, hence leaving PGE2 synthesis largely intact in major parts of the brain while significantly reducing it in the region critical for the febrile response. Furthermore, injection of a virus vector expressing microsomal prostaglandin E synthase-1 (mPGES-1) into the median preoptic nucleus of fever-refractive mPGES-1 knock-out mice, resulted in a temperature elevation in response to LPS. We conclude that the febrile response is dependent on local release of PGE2 onto its target neurons and not on the overall PGE2 production in the brain.SIGNIFICANCE STATEMENT By using mice with selective deletion of prostaglandin synthesis in brain endothelial cells, we demonstrate that local prostaglandin E2 (PGE2) production in deep brain areas, such as the hypothalamus, which is the site of thermoregulatory neurons, is critical for the febrile response to peripheral inflammation. In contrast, PGE2 production in other brain areas and the overall PGE2 level in the brain do not influence the febrile response. Furthermore, partly restoring the PGE2 synthesizing capacity in the anterior hypothalamus of mice lacking such capacity with a lentiviral vector resulted in a temperature elevation in response to LPS. These data imply that the febrile response is dependent on the local release of PGE2 onto its target neurons, possibly by a paracrine mechanism.


Subject(s)
Body Temperature Regulation/immunology , Dinoprostone/biosynthesis , Dinoprostone/immunology , Fever/immunology , Hypothalamus/immunology , Inflammation/immunology , Animals , Female , Fever/etiology , Inflammation/complications , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Hum Mol Genet ; 26(5): 969-988, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28073925

ABSTRACT

Progranulin (PGRN) haploinsufficiency resulting from loss-of-function mutations in the PGRN gene causes frontotemporal lobar degeneration accompanied by TDP-43 accumulation, and patients with homozygous mutations in the PGRN gene present with neuronal ceroid lipofuscinosis. Although it remains unknown why PGRN deficiency causes neurodegenerative diseases, there is increasing evidence that PGRN is implicated in lysosomal functions. Here, we show PGRN is a secretory lysosomal protein that regulates lysosomal function and biogenesis by controlling the acidification of lysosomes. PGRN gene expression and protein levels increased concomitantly with the increase of lysosomal biogenesis induced by lysosome alkalizers or serum starvation. Down-regulation or insufficiency of PGRN led to the increased lysosomal gene expression and protein levels, while PGRN overexpression led to the decreased lysosomal gene expression and protein levels. In particular, the level of mature cathepsin D (CTSDmat) dramatically changed depending upon PGRN levels. The acidification of lysosomes was facilitated in cells transfected with PGRN. Then, this caused degradation of CTSDmat by cathepsin B. Secreted PGRN is incorporated into cells via sortilin or cation-independent mannose 6-phosphate receptor, and facilitated the acidification of lysosomes and degradation of CTSDmat. Moreover, the change of PGRN levels led to a cell-type-specific increase of insoluble TDP-43. In the brain tissue of FTLD-TDP patients with PGRN deficiency, CTSD and phosphorylated TDP-43 accumulated in neurons. Our study provides new insights into the physiological function of PGRN and the role of PGRN insufficiency in the pathogenesis of neurodegenerative diseases.


Subject(s)
Cathepsin D/genetics , DNA-Binding Proteins/genetics , Frontotemporal Lobar Degeneration/genetics , Intercellular Signaling Peptides and Proteins/genetics , Neurons/metabolism , Animals , Brain/metabolism , Brain/pathology , Cathepsin D/metabolism , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Gene Expression Regulation , Haploinsufficiency/genetics , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Lysosomes/genetics , Lysosomes/pathology , Mice , Mutation , Neuroblastoma/metabolism , Neurons/pathology , Primary Cell Culture , Progranulins , Proteins/genetics
20.
Mol Neurobiol ; 54(3): 1953-1966, 2017 04.
Article in English | MEDLINE | ID: mdl-26910812

ABSTRACT

Stress and glucocorticoids suppress adult neurogenesis in the hippocampus. However, the molecular mechanisms underlying stress-induced impairment of adult neurogenesis are poorly understood. We previously suggested that cyclooxygenase (COX)-2 is a common mediator of stresses in the brain. Here, using a lipopolysaccharide (LPS)-induced acute infectious stress model, we evaluated the roles of COX-2 and its major downstream product prostaglandin E2 (PGE2) in adult neurogenesis and the influence of glucocorticoids on COX-2-related signaling. Treatment of rats with LPS significantly decreased neurogenesis in the dentate gyrus (DG) of the hippocampus, and this inhibitory effect of LPS on neurogenesis was reversed by the glucocorticoid receptor antagonist RU486. Moreover, RU486 significantly enhanced the increase in messenger RNA (mRNA) levels of COX-2 and microsomal prostaglandin E synthase (mPGES)-1 in the hippocampus following LPS stimulation. Administration of AH6809, a selective antagonist of the PGE2 EP2 receptor, as well as NS398, a COX-2 selective inhibitor, exacerbated the suppression of proliferation of neural progenitor cells (NPCs) in the DG. Gene expression of EP1, EP2, and EP3, but not EP4, receptors was also increased following LPS stimulation. Immunohistochemical studies indicated that NPCs expressed EP2 receptor, whereas the majority of cells expressing COX-2 and mPGES-1 were mature neurons in the DG. These results suggest that acute infectious stress upregulates COX-2-related signaling in neurons in the DG, which plays a protective role in neurogenesis through EP2 receptor at least partially. In addition, LPS-induced glucocorticoids suppress this COX-2-related signaling, resulting in decreased neurogenesis.


Subject(s)
Communicable Diseases/metabolism , Cyclooxygenase 2/biosynthesis , Glucocorticoids/metabolism , Hippocampus/metabolism , Immunity, Cellular/physiology , Neurogenesis/physiology , Acute Disease , Animals , Communicable Diseases/chemically induced , Communicable Diseases/immunology , Glucocorticoids/antagonists & inhibitors , Glucocorticoids/immunology , Hippocampus/drug effects , Hippocampus/immunology , Immunity, Cellular/drug effects , Lipopolysaccharides/toxicity , Male , Mifepristone/pharmacology , Neurogenesis/drug effects , Rats , Rats, Wistar , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/immunology , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Xanthones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...