Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987435

ABSTRACT

The cerebellum, a phylogenetically ancient brain region, has long been considered strictly a motor control structure. Recent studies have implicated the cerebellum in cognition, sensation, emotion and autonomic function, making it an important target for further investigation. Here, we show that cerebellar Purkinje neurons in mice are activated by the hormone asprosin, leading to enhanced thirst, and that optogenetic or chemogenetic activation of Purkinje neurons induces rapid manifestation of water drinking. Purkinje neuron-specific asprosin receptor (Ptprd) deletion results in reduced water intake without affecting food intake and abolishes asprosin's dipsogenic effect. Purkinje neuron-mediated motor learning and coordination were unaffected by these manipulations, indicating independent control of two divergent functions by Purkinje neurons. Our results show that the cerebellum is a thirst-modulating brain area and that asprosin-Ptprd signaling may be a potential therapeutic target for the management of thirst disorders.

2.
Development ; 149(23)2022 12 01.
Article in English | MEDLINE | ID: mdl-36448532

ABSTRACT

Undescended testis (UDT) affects 6% of male births. Despite surgical correction, some men with unilateral UDT may experience infertility with the contralateral descended testis (CDT) showing no A-dark spermatogonia. To improve our understanding of the etiology of infertility in UDT, we generated a novel murine model of left unilateral UDT. Gubernaculum-specific Wnt4 knockout (KO) mice (Wnt4-cKO) were generated using retinoic acid receptor ß2-cre mice and were found to have a smaller left-unilateral UDT. Wnt4-cKO mice with abdominal UDT had an increase in serum follicle-stimulating hormone and luteinizing hormone and an absence of germ cells in the undescended testicle. Wnt4-cKO mice with inguinal UDT had normal hormonal profiles, and 50% of these mice had no sperm in the left epididymis. Wnt4-cKO mice had fertility defects and produced 52% fewer litters and 78% fewer pups than control mice. Wnt4-cKO testes demonstrated increased expression of estrogen receptor α and SOX9, upregulation of female gonadal genes, and a decrease in male gonadal genes in both CDT and UDT. Several WNT4 variants were identified in boys with UDT. The presence of UDT and fertility defects in Wnt4-cKO mice highlights the crucial role of WNT4 in testicular development.


Subject(s)
Cryptorchidism , Infertility , Female , Male , Humans , Mice , Animals , Gubernaculum , Cryptorchidism/genetics , Fertility/genetics , Spermatogonia , Mice, Knockout , Wnt4 Protein/genetics
3.
Cell Metab ; 34(4): 549-563.e8, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35298903

ABSTRACT

Asprosin is a fasting-induced glucogenic and centrally acting orexigenic hormone. The olfactory receptor Olfr734 is known to be the hepatic receptor for asprosin that mediates its effects on glucose production, but the receptor for asprosin's orexigenic function has been unclear. Here, we have identified protein tyrosine phosphatase receptor δ (Ptprd) as the orexigenic receptor for asprosin. Asprosin functions as a high-affinity Ptprd ligand in hypothalamic AgRP neurons, regulating the activity of this circuit in a cell-autonomous manner. Genetic ablation of Ptprd results in a strong loss of appetite, leanness, and an inability to respond to the orexigenic effects of asprosin. Ablation of Ptprd specifically in AgRP neurons causes resistance to diet-induced obesity. Introduction of the soluble Ptprd ligand-binding domain in the circulation of mice suppresses appetite and blood glucose levels by sequestering plasma asprosin. Identification of Ptprd as the orexigenic asprosin receptor creates a new avenue for the development of anti-obesity therapeutics.


Subject(s)
Peptide Hormones , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Agouti-Related Protein , Animals , Fibrillin-1/metabolism , Glucose/metabolism , Ligands , Mice , Obesity/metabolism , Peptide Fragments/metabolism , Peptide Hormones/genetics , Peptide Hormones/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism
4.
Development ; 148(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33913480

ABSTRACT

Multiple morphological abnormalities of the sperm flagella (MMAF) are a major cause of asthenoteratozoospermia. We have identified protease serine 50 (PRSS50) as having a crucial role in sperm development, because Prss50-null mice presented with impaired fertility and sperm tail abnormalities. PRSS50 could also be involved in centrosome function because these mice showed a threefold increase in acephalic sperm (head-tail junction defect), sperm with multiple heads (spermatid division defect) and sperm with multiple tails, including novel two conjoined sperm (complete or partial parts of several flagellum on the same plasma membrane). Our data support that, in the testis, as in tumorigenesis, PRSS50 activates NFκB target genes, such as the centromere protein leucine-rich repeats and WD repeat domain-containing protein 1 (LRWD1), which is required for heterochromatin maintenance. Prss50-null testes have increased IκκB, and reduced LRWD1 and histone expression. Low levels of de-repressed histone markers, such as H3K9me3, in the Prss50-null mouse testis may cause increases in post-meiosis proteins, such as AKAP4, affecting sperm formation. We provide important insights into the complex mechanisms of sperm development, the importance of testis proteases in fertility and a novel mechanism for MMAF.


Subject(s)
Fertility , Serine Proteases/metabolism , Sperm Tail/enzymology , Testis/enzymology , Animals , Asthenozoospermia/enzymology , Asthenozoospermia/genetics , Heterochromatin/enzymology , Heterochromatin/genetics , Histones/biosynthesis , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Male , Mice , Mice, Mutant Strains , Microtubule Proteins/genetics , Microtubule Proteins/metabolism , Serine Proteases/deficiency , Sperm Head/enzymology
5.
Development ; 148(1)2021 01 13.
Article in English | MEDLINE | ID: mdl-33441379

ABSTRACT

Cryptorchidism is the most common urologic birth defect in men and is a predisposing factor of male infertility and testicular cancer, yet the etiology remains largely unknown. E2F1 microdeletions and microduplications contribute to cryptorchidism, infertility and testicular tumors. Although E2f1 deletion or overexpression in mice causes spermatogenic failure, the mechanism by which E2f1 influences testicular function is unknown. This investigation revealed that E2f1-null mice develop cryptorchidism with severe gubernacular defects and progressive loss of germ cells resulting in infertility and, in rare cases, testicular tumors. It was hypothesized that germ cell depletion resulted from an increase in WNT4 levels. To test this hypothesis, the phenotype of a double-null mouse model lacking both Wnt4 and E2f1 in germ cells was analyzed. Double-null mice are fertile. This finding indicates that germ cell maintenance is dependent on E2f1 repression of Wnt4, supporting a role for Wnt4 in germ cell survival. In the future, modulation of WNT4 expression in men with cryptorchidism and spermatogenic failure due to E2F1 copy number variations may provide a novel approach to improve their spermatogenesis and perhaps their fertility potential after orchidopexy.


Subject(s)
E2F1 Transcription Factor/metabolism , Spermatogenesis , Testis/metabolism , Wnt4 Protein/metabolism , Aging/pathology , Animals , Animals, Newborn , Blood-Testis Barrier/pathology , Cell Cycle/genetics , Cryptorchidism/genetics , Cryptorchidism/pathology , E2F1 Transcription Factor/deficiency , Fertility , Gene Expression Regulation , Male , Mice, Inbred C57BL , Models, Biological , Signal Transduction , Spermatozoa/metabolism , Testis/pathology
6.
Andrology ; 8(5): 1243-1255, 2020 09.
Article in English | MEDLINE | ID: mdl-32385972

ABSTRACT

BACKGROUND: Genitourinary anomalies occur in approximately 1% of humans, but in most cases, the cause is unknown. Aristaless-like homeobox 4 (ALX4) is an important homeodomain transcription factor. ALX4 mutations in humans and mouse have been associated with craniofacial defects and genitourinary anomalies such as cryptorchidism and epispadias. OBJECTIVES: To investigate the presence and the functional impact of ALX4 variants in patients with genitourinary defects. MATERIALS AND METHODS: Two separate patient cohorts were analyzed. One includes clinical exome-sequencing (ES) data from 7500 individuals. The other includes 52 ALX4 Sanger-sequenced individuals with bladder exstrophy-epispadias complex (BEEC). Dual luciferase assays were conducted to investigate the functional transcriptional impact of ALX4 variants in HeLa cells and HEK293 cells. RESULTS: A total of 41 distinct ALX4 heterozygous missense variants were identified in the ES cohort with 15 variants present as recurrent in multiple patients. p.G369E and p.L373F were the only two present in individuals with genitourinary defects. A p.L373F heterozygous variant was also identified in one of the 52 individuals in the BEEC cohort. p.L373F and p.G369E were tested in vitro as both are considered damaging by MutationTaster, although only p.G369E was considered damaging by PolyPhen-2. p.L373F did not alter transcriptional activity in HeLa and HEK293 cells. p.G369E caused a significant 3.4- and 1.8-fold decrease in transcriptional activities relative to wild-type ALX4 in HEK293 and HeLa cells, respectively. DISCUSSION AND CONCLUSIONS: Our study supports the idea that transcription factors like ALX4 could influence the normal development of the GU tract in humans as demonstrated in mouse models as ALX4 variant p.G369E (predicted pathogenic by multiple databases) affects ALX4 function in vitro. Variant p.L373F (predicted pathogenic by only MutationTaster) did not affect ALX4 function in vitro. Exon-sequence information and mouse genetics provide important insights into the complex mechanisms driving genitourinary defects allowing the association of transcriptional defects with congenital disorders.


Subject(s)
DNA-Binding Proteins/genetics , Transcription Factors/genetics , Urogenital Abnormalities/genetics , Genetic Variation , HEK293 Cells , HeLa Cells , Humans
7.
Nat Med ; 23(12): 1444-1453, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29106398

ABSTRACT

Asprosin is a recently discovered fasting-induced hormone that promotes hepatic glucose production. Here we demonstrate that asprosin in the circulation crosses the blood-brain barrier and directly activates orexigenic AgRP+ neurons via a cAMP-dependent pathway. This signaling results in inhibition of downstream anorexigenic proopiomelanocortin (POMC)-positive neurons in a GABA-dependent manner, which then leads to appetite stimulation and a drive to accumulate adiposity and body weight. In humans, a genetic deficiency in asprosin causes a syndrome characterized by low appetite and extreme leanness; this is phenocopied by mice carrying similar mutations and can be fully rescued by asprosin. Furthermore, we found that obese humans and mice had pathologically elevated concentrations of circulating asprosin, and neutralization of asprosin in the blood with a monoclonal antibody reduced appetite and body weight in obese mice, in addition to improving their glycemic profile. Thus, in addition to performing a glucogenic function, asprosin is a centrally acting orexigenic hormone that is a potential therapeutic target in the treatment of both obesity and diabetes.


Subject(s)
Appetite Regulation/genetics , Hypothalamus/metabolism , Microfilament Proteins/physiology , Peptide Fragments/physiology , Peptide Hormones/physiology , Adolescent , Adult , Animals , Appetite Depressants/metabolism , Female , Fibrillin-1 , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/genetics , Neurons/metabolism , Peptide Fragments/genetics , Peptide Hormones/genetics , Rats , Signal Transduction , Young Adult
8.
J Pediatr Adolesc Gynecol ; 30(5): 571-577, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28456695

ABSTRACT

STUDY OBJECTIVE: Paratubal cysts (PTCs) occur in 7%-10% of women, regardless of age. Although common, PTCs often are found incidentally because of the potential for these cysts to be asymptomatic. The specific aims of the study were to determine if PTC number and size correlated with signs of hyperandrogenism and obesity, as well as to investigate the molecular profiles of these PTCs in samples derived from female adolescents. DESIGN, SETTING, PARTICIPANTS, INTERVENTIONS, AND MAIN OUTCOME MEASURES: A prospective cohort study was performed in a single children's hospital. Girls 18 years of age or younger who underwent surgery for PTC suspected on the basis of the presence of a persistent adnexal cyst on imaging or a concern for adnexal torsion involving a cyst were consented to participate in the study. RESULTS: Nineteen patients met enrollment criteria with a mean age at menarche of 11.2 ± 1.3 years. Most of the patients (84%; n = 16/19) had adnexal torsion at the time of diagnosis of PTC. Irregular menses and hirsutism was found in 52.6% (n = 10/19) of the patients, among whom 36.8% (n = 7/19) were obese. The mean PTC size was 10.4 ± 4.3 cm with 57.9% (n = 11/19) of the cohort having more than 1 PTC. When patients were compared on the basis of their body mass index, the size of PTCs was significantly larger in the overweight/obese group. The wingless-type (WNT) signaling members catenin beta 1 (CTNBB1) and wingless-type MMTV integration site family, member 7A (WNT7A) were upregulated in 86% (n = 12/14) and 79% (n = 11/14) of the patients, respectively. WNT7A was significantly upregulated in girls with 1 cyst and low body mass index. CONCLUSION: A correlation exists between obesity, cyst size, and hyperandrogenism. Activation of the WNT/CTNBB1 pathway via WNT7A might play a role in PTC development.


Subject(s)
Hyperandrogenism/complications , Obesity/complications , Parovarian Cyst/complications , Wnt Proteins/metabolism , Adolescent , Adult , Child , Cohort Studies , Female , Hirsutism , Hospitals, Pediatric , Humans , Menarche , Parovarian Cyst/metabolism , Parovarian Cyst/surgery , Prospective Studies , Wnt Signaling Pathway
9.
Methods Mol Biol ; 1344: 205-17, 2016.
Article in English | MEDLINE | ID: mdl-26520126

ABSTRACT

A variety of TGF-ß superfamily members affect adipocyte differentiation and function with consequential effects on energy metabolism. There has been a growing interest in this area because of the apparent influence of the BMP subgroup on brown adipose characteristics and potential application to the treatment of human obesity. In this chapter we describe methods that are useful in allowing one to assess the roles of specific members of the superfamily on adipocyte differentiation and mature adipocyte function, including the isolation and differentiation of mouse embryo fibroblasts (MEFs) to examine cell autonomous effects and the efficient transfection of two commonly used (but difficult to transfect) adipogenic cell lines, C3H/10T1/2 and 3T3-L1.


Subject(s)
Adipogenesis/genetics , Cell Differentiation , Multigene Family , Transforming Growth Factor beta/genetics , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Line , Electroporation , Fibroblasts , Gene Expression , Mice , Plasmids/genetics , Transforming Growth Factor beta/metabolism
11.
J Clin Endocrinol Metab ; 96(4): E674-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21252244

ABSTRACT

CONTEXT: The pseudoautosomal regions (PARs) of the Y-chromosome undergo meiotic recombination with the X-chromosome. PAR mutations are associated with infertility and mental and stature disorders. OBJECTIVE: The aim of the study was to determine whether men with Y-chromosome microdeletions have structural defects in PARs. DESIGN AND PARTICIPANTS: Eighty-seven infertile men with Y-chromosome microdeletions and 35 controls were evaluated for chromosomal rearrangements using commercial or custom (X- and Y-chromosome) array comparative genomic hybridization or by quantitative PCR of selected PAR genes. Multisoftware-defined chromosomal gains or losses were validated by quantitative PCR and FISH. RESULTS: Array comparative genomic hybridization confirmed the AZF deletions identified by multiplex PCR. All men with Y-chromosome microdeletions and an abnormal karyotype displayed PAR abnormalities, as did 10% of men with Y-chromosome microdeletions and a normal karyotype. None of the control subjects or infertile men without Y-chromosome microdeletions had PAR duplications or deletions. SHOX aberrations occurred in 14 men (nine gains and five losses); four were short in stature (<10th percentile), and one was tall (>95th percentile). In contrast, the height of 23 men with Y-chromosome microdeletions and normal PARs was average at 176.8 cm (50th percentile). CONCLUSIONS: Y-chromosome microdeletions can include PAR defects causing genomic disorders such as SHOX, which may be transmitted to offspring. Previously unrecognized PAR gains and losses in men with Y-chromosome microdeletions may have consequences for offspring.


Subject(s)
Chromosome Aberrations , Infertility, Male/genetics , Pseudogenes/genetics , Adult , Body Height/genetics , Case-Control Studies , Chromosome Deletion , Chromosomes, Human, Y/genetics , Comparative Genomic Hybridization , DNA Copy Number Variations , Homeodomain Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Sex Chromosome Aberrations , Sex Chromosome Disorders of Sex Development/genetics , Short Stature Homeobox Protein
12.
Brain ; 134(Pt 4): 1140-55, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21115466

ABSTRACT

Scar tissue at sites of traumatic injury in the adult central nervous system presents a combined physical and molecular impediment to axon regeneration. Of multiple known central nervous system scar associated axon growth inhibitors, semaphorin 3A has been shown to be strongly expressed by invading leptomeningeal fibroblasts. We have previously demonstrated that infusion of the small leucine-rich proteoglycan decorin results in major suppression of several growth inhibitory chondroitin sulphate proteoglycans and growth of adult sensory axons across acute spinal cord injuries. Furthermore, decorin treatment of leptomeningeal fibroblasts significantly increases their ability to support neurite growth of co-cultured adult dorsal root ganglion neurons. In the present study we show that decorin has the ability to suppress semaphorin 3A expression within adult rat cerebral cortex scar tissue and in primary leptomeningeal fibroblasts in vitro. Infusion of decorin core protein for eight days resulted in a significant reduction of semaphorin 3A messenger RNA expression within injury sites compared with saline-treated control animals. Both in situ hybridization and immunostaining confirmed that semaphorin 3A messenger RNA expression and protein levels are significantly reduced in decorin-treated animals. Similarly, decorin treatment decreased the expression of semaphorin 3A messenger RNA in cultured rat leptomeningeal fibroblasts compared with untreated cells. Mechanistic studies revealed that decorin-mediated suppression of semaphorin 3A critically depends on erythroblastic leukaemia viral oncogene homologue B4 and signal transducer and activator of transcription 3 function. Collectively, our studies show that in addition to suppressing the levels of inhibitory chondroitin sulphate proteoglycans, decorin has the ability to suppress semaphorin 3A in the injured central nervous system. Our findings provide further evidence for the use of decorin as a potential therapy for promoting axonal growth and repair in the injured adult mammalian brain and spinal cord.


Subject(s)
Cerebral Cortex/metabolism , Cicatrix/metabolism , Decorin/metabolism , ErbB Receptors/metabolism , STAT3 Transcription Factor/metabolism , Semaphorin-3A/metabolism , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Decorin/pharmacology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Immunohistochemistry , In Situ Hybridization , Nerve Regeneration/physiology , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley , Receptor, ErbB-4 , Reverse Transcriptase Polymerase Chain Reaction
13.
Curr Opin Endocrinol Diabetes Obes ; 17(5): 446-52, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20585248

ABSTRACT

PURPOSE OF REVIEW: The review highlights recent findings regarding the functions of mitochondria in adipocytes, providing an understanding of their central roles in regulating substrate metabolism, energy expenditure, disposal of reactive oxygen species (ROS), and in the pathophysiology of obesity and insulin resistance, as well as roles in the mechanisms that affect adipogenesis and mature adipocyte function. RECENT FINDINGS: Nutrient excess leads to mitochondrial dysfunction, which in turn leads to obesity-related pathologies, in part due to the harmful effects of ROS. The recent recognition of 'ectopic' brown adipose in humans suggests that this tissue may play an underappreciated role in the control of energy expenditure. Transcription factors, PGC-1alpha and PRDM16, which regulate brown adipogenesis, and members of the TGF-beta superfamily that modulate this process may be important new targets for antiobesity drugs. SUMMARY: Mitochondria play central roles in ATP production, energy expenditure, and disposal of ROS. Excessive energy substrates lead to mitochondrial dysfunction with consequential effects on lipid and glucose metabolism. Adipocytes help to maintain the appropriate balance between energy storage and expenditure and maintaining this balance requires normal mitochondrial function. Many adipokines, including members of the TGF-beta superfamily, and transcriptional coactivators, PGC-1alpha and PRDM16, are important regulators of this process.


Subject(s)
Adipose Tissue/metabolism , Mitochondrial Diseases/metabolism , Obesity/metabolism , Adipogenesis/physiology , Adipokines/metabolism , Adipose Tissue/physiopathology , Animals , DNA-Binding Proteins/metabolism , Energy Metabolism , Glucose/metabolism , Heat-Shock Proteins/metabolism , Humans , Insulin Resistance/physiology , Ion Channels/metabolism , Lipid Metabolism/physiology , Mice , Mitochondrial Proteins/metabolism , Obesity/physiopathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Uncoupling Protein 1
14.
Endocrinology ; 150(8): 3521-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19389832

ABSTRACT

Activin-betaA and activin-betaB (encoded by Inhba and Inhbb genes, respectively) are closely related TGF-beta superfamily members that participate in a variety of biological processes. We previously generated mice with an insertion allele at the Inhba locus, Inhba(BK). In this allele, the sequence encoding the Inhba mature domain is replaced with that of Inhbb, rendering the gene product functionally hypomorphic. Homozygous (Inhba(BK/BK)) and hemizygous (Inhba(BK/-)) mice are smaller and leaner than their wild-type littermates, and many tissues are disproportionately small relative to total body weight. To determine the mechanisms that contribute to these phenomena, we investigated the metabolic consequences of the mutation. Although the growth of Inhba(BK) mice is improved by providing a calorie-rich diet, diet-induced obesity, fatty liver, and insulin resistance (hallmarks of chronic caloric excess) do not develop, despite greater caloric intake than wild-type controls. Physiological, molecular, and biochemical analyses all revealed characteristics that are commonly associated with increased mitochondrial energy metabolism, with a corresponding up-regulation of several genes that reflect enhanced mitochondrial biogenesis and function. Oxygen consumption, an indirect measure of the metabolic rate, was markedly increased in Inhba(BK/BK) mice, and polarographic analysis of liver mitochondria revealed an increase in ADP-independent oxygen consumption, consistent with constitutive uncoupling of the inner mitochondrial membrane. These findings establish a functional relationship between activin signaling and mitochondrial energy metabolism and further support the rationale to target this signaling pathway for the medical treatment of cachexia, obesity, and diabetes.


Subject(s)
Body Composition/genetics , Energy Metabolism/genetics , Inhibin-beta Subunits/physiology , Animals , Blood Glucose/drug effects , Blood Glucose/genetics , Body Weight/genetics , Calorimetry, Indirect , Dietary Fats/pharmacology , Energy Intake/genetics , Enzyme-Linked Immunosorbent Assay , Female , Glucose Tolerance Test , In Vitro Techniques , Inhibin-beta Subunits/genetics , Insulin Resistance/genetics , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Mice , Mice, Mutant Strains , Organ Size/genetics , Oxygen Consumption , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction
15.
J Neurotrauma ; 23(3-4): 397-408, 2006.
Article in English | MEDLINE | ID: mdl-16629625

ABSTRACT

Spinal cord scar tissue presents a combined physical and molecular barrier to axon regeneration. Theoretically, spinal cord injuries (SCIs) can be rendered more permissive to axon growth by either suppressing synthesis of misaligned, fibrotic scar tissue and associated axon growth inhibitors, or enzymatically degrading them. We have previously shown that acute infusion of human recombinant decorin core protein into discreet stab injuries of the rat dorsal column pathways effected a major suppression of inflammation, astrogliosis, and multiple axon growth inhibitory chondroitin sulfate proteoglycans, which combined to promote rapid axon growth across the injury site. The high efficiency of chondroitin sulfate proteoglycan (CSPG) core protein suppression (approximately 90%) suggested that decorin may promote CSPG degradation in addition to suppressing CSPG synthesis. As the serine protease plasmin can degrade axon growth inhibitory CSPGs (neurocan and phosphacan) and its zymogen, plasmininogen is synthesized by microglia, we have investigated whether decorin treatment of acute SCIs and cultured adult spinal cord microglia can increase plasminogen/ plasmin synthesis. Infusion of hr-decorin over the first 8 days post-SCI induced 10- and 17-fold increases in plasminogen and plasmin protein levels, respectively, within sites of injury and a threefold increase in microglial plasminogen mRNA in vitro. In addition to potentially degrading multiple axon growth inhibitory components of the glial scar, plasmin is known to play major roles in activating neurotrophins and promoting central nervous system (CNS) plasticity. The wider implications of decorin induction of plasmin in the injured spinal cord for axon regeneration, and recovery of function at acute and chronic time points post-SCI are reviewed.


Subject(s)
Extracellular Matrix Proteins/pharmacology , Fibrinolysin/biosynthesis , Microglia/metabolism , Plasminogen/biosynthesis , Proteoglycans/pharmacology , Spinal Cord Injuries/metabolism , Animals , Animals, Genetically Modified , Blotting, Western , Cicatrix/pathology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Decorin , Female , Mammary Glands, Animal/metabolism , Mice , Microglia/drug effects , Nerve Regeneration/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL