Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Autophagy ; 18(5): 1127-1151, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35287553

RESUMO

Parkinson disease (PD) is a neurodegenerative disorder characterized by the abnormal intracellular accumulation of SNCA/α-synuclein. While the exact mechanisms underlying SNCA pathology are not fully understood, increasing evidence suggests the involvement of autophagy as well as lysosomal deficiencies. Because CTSD (cathepsin D) has been proposed to be the major lysosomal protease involved in SNCA degradation, its deficiency has been linked to the presence of insoluble SNCA conformers in the brain of mice and humans as well as to the transcellular transmission of SNCA aggregates. We here postulate that SNCA degradation can be enhanced by the application of the recombinant human proform of CTSD (rHsCTSD). Our results reveal that rHsCTSD is efficiently endocytosed by neuronal cells, correctly targeted to lysosomes and matured to an enzymatically active protease. In dopaminergic neurons derived from induced pluripotent stem cells (iPSC) of PD patients harboring the A53T mutation within the SNCA gene, we confirm the reduction of insoluble SNCA after treatment with rHsCTSD. Moreover, we demonstrate a decrease of pathological SNCA conformers in the brain and within primary neurons of a ctsd-deficient mouse model after dosing with rHsCTSD. Boosting lysosomal CTSD activity not only enhanced SNCA clearance in human and murine neurons as well as tissue, but also restored endo-lysosome and autophagy function. Our findings indicate that CTSD is critical for SNCA clearance and function. Thus, enzyme replacement strategies utilizing CTSD may also be of therapeutic interest for the treatment of PD and other synucleinopathies aiming to decrease the SNCA burden.Abbreviations: aa: amino acid; SNCA/α-synuclein: synuclein alpha; APP: amyloid beta precursor protein; BBB: blood brain barrier; BF: basal forebrain; CBB: Coomassie Brilliant Blue; CLN: neuronal ceroid lipofuscinosis; CNL10: neuronal ceroid lipofuscinosis type 10; Corr.: corrected; CTSD: cathepsin D; CTSB: cathepsin B; DA: dopaminergic; DA-iPSn: induced pluripotent stem cell-derived dopaminergic neurons; dox: doxycycline; ERT: enzyme replacement therapy; Fx: fornix, GBA/ß-glucocerebrosidase: glucosylceramidase beta; h: hour; HC: hippocampus; HT: hypothalamus; i.c.: intracranially; IF: immunofluorescence; iPSC: induced pluripotent stem cell; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LSDs: lysosomal storage disorders; MAPT: microtubule associated protein tau; M6P: mannose-6-phosphate; M6PR: mannose-6-phosphate receptor; MB: midbrain; mCTSD: mature form of CTSD; neurofil.: neurofilament; PD: Parkinson disease; proCTSD: proform of CTSD; PRNP: prion protein; RFU: relative fluorescence units; rHsCTSD: recombinant human proCTSD; SAPC: Saposin C; SIM: structured illumination microscopy; T-insol: Triton-insoluble; T-sol: Triton-soluble; TEM: transmission electron microscopy, TH: tyrosine hydroxylase; Thal: thalamus.


Assuntos
Lipofuscinoses Ceroides Neuronais , Doença de Parkinson , Sinucleinopatias , Peptídeos beta-Amiloides/metabolismo , Animais , Autofagia/fisiologia , Catepsina D/deficiência , Catepsina D/metabolismo , Neurônios Dopaminérgicos/metabolismo , Humanos , Lisossomos/metabolismo , Camundongos , Lipofuscinoses Ceroides Neuronais/metabolismo , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo
3.
Theranostics ; 11(13): 6173-6192, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995652

RESUMO

Rationale: Alternative therapeutic strategies based on tumor-specific molecular targets are urgently needed for triple-negative breast cancer (TNBC). The protease cathepsin D (cath-D) is a marker of poor prognosis in TNBC and a tumor-specific extracellular target for antibody-based therapy. The identification of cath-D substrates is crucial for the mechanistic understanding of its role in the TNBC microenvironment and future therapeutic developments. Methods: The cath-D substrate repertoire was investigated by N-Terminal Amine Isotopic Labeling of Substrates (TAILS)-based degradome analysis in a co-culture assay of TNBC cells and breast fibroblasts. Substrates were validated by amino-terminal oriented mass spectrometry of substrates (ATOMS). Cath-D and SPARC expression in TNBC was examined using an online transcriptomic survival analysis, tissue micro-arrays, TNBC cell lines, patient-derived xenografts (PDX), human TNBC samples, and mammary tumors from MMTV-PyMT Ctsd-/- knock-out mice. The biological role of SPARC and its fragments in TNBC were studied using immunohistochemistry and immunofluorescence analysis, gene expression knockdown, co-culture assays, western blot analysis, RT-quantitative PCR, adhesion assays, Transwell motility, trans-endothelial migration and invasion assays. Results: TAILS analysis showed that the matricellular protein SPARC is a substrate of extracellular cath-D. In vitro, cath-D induced limited proteolysis of SPARC C-terminal extracellular Ca2+ binding domain at acidic pH, leading to the production of SPARC fragments (34-, 27-, 16-, 9-, and 6-kDa). Similarly, cath-D secreted by TNBC cells cleaved fibroblast- and cancer cell-derived SPARC at the tumor pericellular acidic pH. SPARC cleavage also occurred in TNBC tumors. Among these fragments, only the 9-kDa SPARC fragment inhibited TNBC cell adhesion and spreading on fibronectin, and stimulated their migration, endothelial transmigration, and invasion. Conclusions: Our study establishes a novel crosstalk between proteases and matricellular proteins in the tumor microenvironment through limited SPARC proteolysis, revealing a novel targetable 9-kDa bioactive SPARC fragment for new TNBC treatments. Our study will pave the way for the development of strategies for targeting bioactive fragments from matricellular proteins in TNBC.


Assuntos
Catepsina D/metabolismo , Matriz Extracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Osteonectina/metabolismo , Fragmentos de Peptídeos/farmacologia , Neoplasias de Mama Triplo Negativas/patologia , Microambiente Tumoral , Sequência de Aminoácidos , Animais , Sítios de Ligação , Catepsina D/deficiência , Catepsina D/genética , Adesão Celular , Feminino , Fibroblastos , Regulação Neoplásica da Expressão Gênica , Humanos , Concentração de Íons de Hidrogênio , Neoplasias Mamárias Experimentais/enzimologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Peso Molecular , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Osteonectina/genética , Fragmentos de Peptídeos/metabolismo , Domínios Proteicos , Proteólise , Especificidade por Substrato , Migração Transendotelial e Transepitelial , Neoplasias de Mama Triplo Negativas/enzimologia
4.
Cells ; 10(3)2021 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-33800998

RESUMO

Vision loss is among the characteristic symptoms of neuronal ceroid lipofuscinosis (NCL), a fatal neurodegenerative lysosomal storage disorder. Here, we performed an in-depth analysis of retinal degeneration at the molecular and cellular levels in mice lacking the lysosomal aspartyl protease cathepsin D, an animal model of congenital CLN10 disease. We observed an early-onset accumulation of storage material as indicated by elevated levels of saposin D and subunit C of the mitochondrial ATP synthase. The accumulation of storage material was accompanied by reactive astrogliosis and microgliosis, elevated expression of the autophagy marker sequestosome 1/p62 and a dysregulated expression of several lysosomal proteins. The number of cone photoreceptor cells was reduced as early as at postnatal day 5. At the end stage of the disease, the outer nuclear layer was almost atrophied, and all cones were lost. A significant loss of rod and cone bipolar cells, amacrine cells and ganglion cells was found at advanced stages of the disease. Results demonstrate that cathepsin D deficiency results in an early-onset and rapidly progressing retinal dystrophy that involves all retinal cell types. Data of the present study will serve as a reference for studies aimed at developing treatments for retinal degeneration in CLN10 disease.


Assuntos
Catepsina D/deficiência , Lipofuscinoses Ceroides Neuronais/patologia , Retina/patologia , Animais , Autofagia , Catepsina D/metabolismo , Modelos Animais de Doenças , Gliose/patologia , Lisossomos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Lipofuscinoses Ceroides Neuronais/complicações , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Subunidades Proteicas/metabolismo , Degeneração Retiniana/complicações , Degeneração Retiniana/patologia , Proteína Sequestossoma-1/metabolismo
5.
Sci Rep ; 11(1): 6596, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33758261

RESUMO

The major lysosomal proteases, Cathepsin B (CTSB), Cathepsin D (CTSD) and Cathepsin L (CTSL), are implicated in autophagic activity. To investigate the role of each cathepsin in the exocrine pancreas, we generated mice in which the pancreas was specifically deficient in Ctsb, Ctsd and Ctsl. Each of these gene knockout (KO) and Ctsb;Ctsl and Ctsd;Ctsl double-knockout (DKO) mice were almost normal. However, we found cytoplasmic degeneration in the pancreatic acinar cells of Ctsb;Ctsd DKO mice, similar to autophagy related 5 (Atg5) KO mice. LC3 and p62 (autophagy markers) showed remarkable accumulation and the numbers of autophagosomes and autolysosomes were increased in the pancreatic acinar cells of Ctsb;Ctsd DKO mice. Moreover, these Ctsb;Ctsd DKO mice also developed chronic pancreatitis (CP). Thus, we conclude that both Ctsb and Ctsd deficiency caused impaired autophagy in the pancreatic acinar cells, and induced CP in mice.


Assuntos
Autofagia , Catepsina B/deficiência , Catepsina D/deficiência , Pâncreas/metabolismo , Pancreatite Crônica/metabolismo , Células Acinares/metabolismo , Animais , Autofagossomos/metabolismo , Catepsina B/genética , Catepsina B/metabolismo , Catepsina D/genética , Catepsina D/metabolismo , Camundongos , Pâncreas/citologia , Pancreatite Crônica/genética
6.
J Mass Spectrom ; 56(1): e4675, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33314470

RESUMO

Neuronal ceroid lipofuscinoses (NCLs) are a group of neurodegenerative diseases predominantly in childhood that are characterized by psychomotor deterioration, epilepsy, and early death of patients. The NCLs analyzed in the present study are caused by defects of the specific enzymes, CLN1 (palmitoyl protein thioesterase 1; PPT1), CLN2 (tripeptidyl peptidase 1; TPP1), and CLN10 (cathepsin D). Specific and sensitive diagnostic assays of NCLs were the main goal of this study. They are of increasing importance, particularly since enzyme replacement therapy (ERT) for NCL2 has recently become available for clinical treatment, and ERTs for further NCLs are under development. Here, we report specific and sensitive determinations for CLN1, CLN2, and CLN10 on dried blood spots by tandem mass spectrometry using multiple reaction monitoring mass spectrometry (MRM-MS). Identical substrates suitable for (i) fluorimetric determination of single enzymes and (ii) for MRM-MS determination of multiple enzymes were synthesized by chemical coupling of alkyl-umbelliferone building blocks with the corresponding peptidyl-substrate groups recognized by the target enzyme. Enzymatic determinations were performed both by fluorimetry and MRM-MS in patients with NCL1, NCL2, and NCL10 and showed good agreement in single assays. Moreover, duplex and triplex determinations were successfully performed for NCL1, NCL2, and NCL10. Specific peptidyl-(4-alkyl-umbelliferone) substrates were also synthesized for mass spectrometric determinations of different cathepsins (cathepsins-D, -F, and -B), to provide a differentiation of proteolytic specificities.


Assuntos
Teste em Amostras de Sangue Seco/métodos , Fluorometria/métodos , Lipofuscinoses Ceroides Neuronais/sangue , Espectrometria de Massas em Tandem/métodos , Adolescente , Catepsina D/sangue , Catepsina D/deficiência , Criança , Pré-Escolar , Humanos , Proteínas de Membrana/sangue , Lipofuscinoses Ceroides Neuronais/diagnóstico , Lipofuscinoses Ceroides Neuronais/enzimologia , Proteínas Nucleares/sangue , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Especificidade por Substrato , Tioléster Hidrolases/sangue , Tripeptidil-Peptidase 1
7.
Nat Commun ; 11(1): 5133, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-33046706

RESUMO

Cathepsin D (CTSD) is a lysosomal protease and a marker of poor prognosis in breast cancer. However, the cells responsible for this association and the function of CTSD in cancer are still incompletely understood. By using a conditional CTSD knockout mouse crossed to the transgenic MMTV-PyMT breast cancer model we demonstrate that CTSD deficiency in the mammary epithelium, but not in myeloid cells, blocked tumor development in a cell-autonomous manner. We show that lack of CTSD impaired mechanistic Target of Rapamycin Complex 1 (mTORC1) signaling and induced reversible cellular quiescence. In line, CTSD-deficient tumors started to grow with a two-month delay and quiescent Ctsd-/- tumor cells re-started proliferation upon long-term culture. This was accompanied by rewiring of oncogenic gene expression and signaling pathways, while mTORC1 signaling remained permanently disabled in CTSD-deficient cells. Together, these studies reveal a tumor cell-autonomous effect of CTSD deficiency, and establish a pivotal role of this protease in the cellular response to oncogenic stimuli.


Assuntos
Neoplasias da Mama/metabolismo , Catepsina D/genética , Epitélio/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Animais , Neoplasias da Mama/genética , Catepsina D/deficiência , Feminino , Humanos , Glândulas Mamárias Animais/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
8.
Int J Mol Sci ; 20(7)2019 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-30959855

RESUMO

Cathepsin D is one of the major lysosomal aspartic proteases that is essential for the normal functioning of the autophagy-lysosomal system. In the kidney, cathepsin D is enriched in renal proximal tubular epithelial cells, and its levels increase during acute kidney injury. To investigate how cathepsin D-deficiency impacts renal proximal tubular cells, we employed a conditional knockout CtsDflox/-; Spink3Cre mouse. Immunohistochemical analyses using anti-cathepsin D antibody revealed that cathepsin D was significantly decreased in tubular epithelial cells of the cortico-medullary region, mainly in renal proximal tubular cells of this mouse. Cathepsin D-deficient renal proximal tubular cells showed an increase of microtubule-associated protein light chain 3 (LC3; a marker for autophagosome/autolysosome)-signals and an accumulation of abnormal autophagic structures. Renal ischemia/reperfusion injury resulted in an increase of early kidney injury marker, Kidney injury molecule 1 (Kim-1), in the cathepsin D-deficient renal tubular epithelial cells of the CtsDflox/-; Spink3Cre mouse. Inflammation marker was also increased in the cortico-medullary region of the CtsDflox/-; Spink3Cre mouse. Our results indicated that lack of cathepsin D in the renal tubular epithelial cells led to an increase of sensitivity against ischemia/reperfusion injury.


Assuntos
Catepsina D/deficiência , Túbulos Renais Proximais/enzimologia , Túbulos Renais Proximais/patologia , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Animais , Autofagia , Catepsina D/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Integrases/metabolismo , Camundongos
9.
Biotechnol Prog ; 35(4): e2820, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30972970

RESUMO

Chinese hamster ovary (CHO) cells have been used as host cells in the production of a range of recombinant therapeutic proteins, including monoclonal antibodies and Fc-fusion proteins. Host cell proteins (HCP) represent impurities that must be removed from therapeutic formulations because of their potential risks for immunogenicity. While the majority of HCP impurities are effectively removed in typical downstream purification processes, clearance of a small population of HCP remains challenging. In this study, we knocked out the Anxa2 and Ctsd genes to assess the feasibility of knockout approaches for diminishing the risk of contamination with HCP. Using the CRISPR/Cas9 system, Anxa2-, and Ctsd-knockout CHO cell lines were successfully established, and we confirmed the complete elimination of the corresponding HCP in cell lysates. Importantly, all knockout cell lines showed similar growth and viability to those of the wild-type control during 8 days of cultivation. Thus, knockout of unrequired genes can reduce contamination with HCP in the production of recombinant therapeutic proteins.


Assuntos
Anexina A2/química , Anticorpos Monoclonais/química , Catepsina D/química , Proteínas Recombinantes de Fusão/química , Animais , Anexina A2/deficiência , Anexina A2/genética , Células CHO , Catepsina D/deficiência , Catepsina D/genética , Sobrevivência Celular , Células Cultivadas , Cricetulus , Humanos
10.
Exp Mol Med ; 50(3): e457, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29546879

RESUMO

This study aimed to investigate the role of cathepsin D (CathD) in central nervous system (CNS) myelination and its possible mechanism. By using CathD knockout mice in conjunction with immunohistochemistry, immunocytochemistry and western blot assays, the myelination of the CNS and the development of oligodendrocyte lineage cells in vivo and in vitro were observed. Endocytosis assays, real-time-lapse experiments and total internal reflection fluorescence microscopy were used to demonstrate the location and movement of proteolipid protein in oligodendrocyte lineage cells. In addition, the relevant molecular mechanism was explored by immunoprecipitation. The increase in Fluoromyelin Green staining and proteolipid protein expression was not significant in the corpus callosum of CathD-/- mice at the age of P11, P14 and P24. Proteolipid protein expression was weak at each time point and was mostly accumulated around the nucleus. The number of oligodendrocyte lineage cells (olig2+) and mature oligodendrocytes (CC1+) significantly decreased between P14 and P24. In the oligodendrocyte precursor cell culture of CathD-/- mice, the morphology of myelin basic protein-positive mature oligodendrocytes was simple while oligodendrocyte precursor cells showed delayed differentiation into mature oligodendrocytes. Moreover, more proteolipid protein gathered in late endosomes/lysosomes (LEs/Ls) and fewer reached the plasma membrane. Immunohistochemistry and immunoelectron microscopy analysis showed that CathD, proteolipid protein and VAMP7 could bind with each other, whereas VAMP7 and proteolipid protein colocalized with CathD in late endosome/lysosome. The findings of this paper suggest that CathD may have an important role in the myelination of CNS, presumably by altering the trafficking of proteolipid protein.


Assuntos
Catepsina D/deficiência , Catepsina D/metabolismo , Membrana Celular/metabolismo , Sistema Nervoso Central/metabolismo , Endossomos/metabolismo , Lisossomos/metabolismo , Proteolipídeos/metabolismo , Animais , Catepsina D/genética , Imunoprecipitação , Camundongos , Camundongos Knockout , Bainha de Mielina/metabolismo
11.
Hum Mol Genet ; 26(24): 4861-4872, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29036611

RESUMO

Frontotemporal dementia (FTD) encompasses a group of neurodegenerative disorders characterized by cognitive and behavioral impairments. Heterozygous mutations in progranulin (PGRN) cause familial FTD and result in decreased PGRN expression, while homozygous mutations result in complete loss of PGRN expression and lead to the neurodegenerative lysosomal storage disorder neuronal ceroid lipofuscinosis (NCL). However, how dose-dependent PGRN mutations contribute to these two different diseases is not well understood. Using iPSC-derived human cortical neurons from FTD patients harboring PGRN mutations, we demonstrate that PGRN mutant neurons exhibit decreased nuclear TDP-43 and increased insoluble TDP-43, as well as enlarged electron-dense vesicles, lipofuscin accumulation, fingerprint-like profiles and granular osmiophilic deposits, suggesting that both FTD and NCL-like pathology are present in PGRN patient neurons as compared to isogenic controls. PGRN mutant neurons also show impaired lysosomal proteolysis and decreased activity of the lysosomal enzyme cathepsin D. Furthermore, we find that PGRN interacts with cathepsin D, and that PGRN increases the activity of cathepsin D but not cathepsins B or L. Finally, we show that granulin E, a cleavage product of PGRN, is sufficient to increase cathepsin D activity. This functional relationship between PGRN and cathepsin D provides a possible explanation for overlapping NCL-like pathology observed in patients with mutations in PGRN or CTSD, the gene encoding cathepsin D. Together, our work identifies PGRN as an activator of lysosomal cathepsin D activity, and suggests that decreased cathepsin D activity due to loss of PGRN contributes to both FTD and NCL pathology in a dose-dependent manner.


Assuntos
Catepsina D/deficiência , Demência Frontotemporal/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Catepsina D/genética , Catepsina D/metabolismo , Linhagem Celular Transformada , Proteínas de Ligação a DNA/genética , Fibroblastos/patologia , Demência Frontotemporal/genética , Demência Frontotemporal/patologia , Células HEK293 , Haploinsuficiência , Heterozigoto , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Lisossomos/metabolismo , Mutação , Lipofuscinoses Ceroides Neuronais/genética , Lipofuscinoses Ceroides Neuronais/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Progranulinas
13.
Biochimie ; 122: 219-26, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26232697

RESUMO

Cathepsin D (Ctsd) is a ubiquitously expressed aspartic protease functioning primarily in the acidic endosomal/lysosomal cell compartment. At an age of 26 ± 1 days, mice with constitutive Ctsd deficiency (Ctsd(-/-)) die from a neurodegenerative lysosomal storage disease equivalent to the congenital neuronal ceroid lipofuscinosis (NCL) type 10 in humans. In addition to neurodegeneration, Ctsd(-/-) mice exhibit a loss of CD4(+)/CD8(+)-double-positive thymocytes and an atrophy of the intestinal mucosa. To date, it is not understood if and how these phenotypes are triggering each other. In addition, the cell type causing initiation of NCL in Ctsd(-/-) mice has not been identified yet. To investigate the tissue- and cell type-specific functions of Ctsd, we generated a novel conditional Ctsd allele by flanking the second exon with loxP sites. We compared a ubiquitous Ctsd deletion with a deletion of the protease by a Nestin-promoter controlled Cre-recombinase expression in cells of neuroectodermal origin, e.g. in neurons and astroglia, but not in microglia. First, we confirmed absence of Ctsd in the respective cell- and tissue types. The neuroectoderm specific knock-out mice survived about 5.5 days longer than the mice with ubiquitous Ctsd deletion, which was in line with the progress in brain histopathology. Atrophies of thymus and small intestine were delayed to similar extend. The conditional Ctsd knock-out mouse model established in this study not only demonstrates that this type of NCL is initiated by cells of neuroectodermal origin, but will also help to further study tissue-specific functions of Ctsd in vivo.


Assuntos
Catepsina D/deficiência , Modelos Animais de Doenças , Ectoderma/metabolismo , Lipofuscinoses Ceroides Neuronais/enzimologia , Animais , Astrócitos/enzimologia , Astrócitos/metabolismo , Atrofia/genética , Western Blotting , Catepsina D/genética , Deleção de Genes , Regulação Enzimológica da Expressão Gênica , Humanos , Imuno-Histoquímica , Intestino Delgado/enzimologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Camundongos Knockout , Tubo Neural/metabolismo , Lipofuscinoses Ceroides Neuronais/genética , Neurônios/enzimologia , Neurônios/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida , Timo/enzimologia , Timo/metabolismo , Timo/patologia , Fatores de Tempo
14.
Hum Mol Genet ; 24(19): 5416-32, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26160911

RESUMO

Neurodegeneration is a devastating manifestation in the majority of >50 lysosomal storage disorders (LSDs). Neuronal ceroid lipofuscinoses (NCLs) are the most common childhood neurodegenerative LSDs. Mutations in 13 different genes (called CLNs) underlie various types of NCLs, of which the infantile NCL (INCL) and congenital NCL (CNCL) are the most lethal. Although inactivating mutations in the CLN1 gene encoding palmitoyl-protein thioesterase-1 (PPT1) cause INCL, those in the CLN10 gene encoding cathepsin D (CD) underlie CNCL. PPT1 is a lysosomal thioesterase that cleaves the thioester linkage in S-acylated proteins required for their degradation by lysosomal hydrolases like CD. Thus, PPT1 deficiency causes lysosomal accumulation of these lipidated proteins (major constituents of ceroid) leading to INCL. We sought to determine whether there is a common pathogenic link between INCL and CNCL. Using biochemical, histological and confocal microscopic analyses of brain tissues and cells from Cln1(-/-) mice that mimic INCL, we uncovered that Cln10/CD is overexpressed. Although synthesized in the endoplasmic reticulum, the CD-precursor protein (pro-CD) is transported through endosome to the lysosome where it is proteolytically processed to enzymatically active-CD. We found that despite Cln10 overexpression, the maturation of pro-CD to enzymatically active-CD in lysosome was disrupted. This defect impaired lysosomal degradative function causing accumulation of undegraded cargo in lysosome leading to INCL. Notably, treatment of intact Cln1(-/-) mice as well as cultured brain cells derived from these animals with a thioesterase-mimetic small molecule, N-tert-butyl-hydroxylamine, ameliorated the CD-processing defect. Our findings are significant in that they define a pathway in which Cln1 mutations disrupt the maturation of a major degradative enzyme in lysosome contributing to neuropathology in INCL and suggest that lysosomal CD deficiency is a common pathogenic link between INCL and CNCL.


Assuntos
Encéfalo/metabolismo , Catepsina D/metabolismo , Lipofuscinoses Ceroides Neuronais/patologia , Tioléster Hidrolases/genética , Animais , Encéfalo/patologia , Catepsina D/deficiência , Criança , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Hidroxilaminas/administração & dosagem , Hidroxilaminas/uso terapêutico , Lisossomos/metabolismo , Camundongos , Mutação , Lipofuscinoses Ceroides Neuronais/tratamento farmacológico , Lipofuscinoses Ceroides Neuronais/genética
15.
Wien Med Wochenschr ; 165(9-10): 210-3, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26059544

RESUMO

BACKGROUND: Neuronal ceroid lipofuscinoses (NCL) is characterized by a combination of retinopathy, dementia, and epilepsy. As a group, they encompass ten distinct biological and clinical entities and are the most common type of childhood neurodegenerative disease. PATIENTS AND METHODS: Case reports. RESULTS: We demonstrate the clinical course of two neonates (brother and sister) with infantile neuronal ceroid lipofuscinoses (NCL) (CLN 10 disease) presenting with intractable seizures and respiratory insufficiency immediately after birth. Characteristic clinical, radiological and pathological findings of this form of NCL are presented. CONCLUSIONS: We conclude that the diagnosis of CLN10 should be kept in mind as a differential diagnosis in newborns presenting with respiratory insufficiency and severe epilepsy that is largely refractory to anti-epileptic drugs (AED) treatment. Because of the severity of CLN10 disease and futility of treatment, important ethical issues arise when caring for children with this clinical entity.


Assuntos
Catepsina D/deficiência , Lipofuscinoses Ceroides Neuronais/genética , Adulto , Encéfalo/anormalidades , Encéfalo/patologia , Catepsina D/genética , Aberrações Cromossômicas , Consanguinidade , Diagnóstico Diferencial , Ética Médica , Eutanásia Passiva/ética , Feminino , Genes Recessivos/genética , Triagem de Portadores Genéticos , Humanos , Recém-Nascido , Masculino , Lipofuscinoses Ceroides Neuronais/diagnóstico , Lipofuscinoses Ceroides Neuronais/terapia , Cuidados Paliativos/ética
16.
Artigo em Inglês | MEDLINE | ID: mdl-26092248

RESUMO

Mutations in cathepsin D (CTSD), an aspartic protease in the endosomal-lysosomal system, underlie congenital neuronal ceroid-lipofuscinosis (cNCL, also known as CLN10), a devastating neurodegenerative disease. CLN10 patients die within the first few days of life, and in the few patients who live into adulthood psychopathological symptoms have not been reported. Extensive neuropathology and altered neurotransmission have been reported in CTSD-deficient mice; however signs of neuropsychiatric behavior in these mice are not well characterized due to the severe movement disorder and premature death of the animal. In the present study, we show that heterozygous CTSD-deficient (CTSD HET) mice display an overall behavioral profile that is similar to human mania, including hyperlocomotion, d-amphetamine-induced hyperactivity, sleep-disturbance, and reduced anxiety-like behavior. However, under stressful conditions CTSD HET mice manifest depressive-like behavior, including anhedonia, behavioral despair, and enhanced learned helplessness. Chronic administration of lithium chloride or valproic acid, two clinically effective mood stabilizers, reverses the majority of these behavioral abnormalities. In addition, CTSD HET mice display stress-induced hypersecretion of corticosterone. These findings suggest an important role for CTSD in the regulation of mood stabilization.


Assuntos
Transtorno Bipolar/etiologia , Transtorno Bipolar/genética , Catepsina D/deficiência , Depressão/etiologia , Depressão/genética , Lipofuscinoses Ceroides Neuronais/complicações , Adaptação Ocular/efeitos dos fármacos , Adaptação Ocular/genética , Animais , Antidepressivos/uso terapêutico , Catepsina D/genética , Corticosterona/sangue , Dextroanfetamina/farmacologia , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Feminino , Preferências Alimentares/efeitos dos fármacos , Hipercinese/tratamento farmacológico , Hipercinese/genética , Cloreto de Lítio/uso terapêutico , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transtornos do Sono-Vigília/etiologia , Natação/psicologia , Ácido Valproico/uso terapêutico
17.
Mol Cell Neurosci ; 64: 51-60, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25496868

RESUMO

Recent evidence suggests that peripheral blood mononuclear cells (PBMCs) contribute to the pathogenesis of neuropathological changes in patients with neuronal ceroid lipofuscinosis (NCL) and lysosomal storage diseases. In order to examine the possible increase in the permeability of the blood-brain-barrier (BBB) and resultant infiltration of PBMCs due to cathepsin D (CatD) deficiency, a process underlying the onset of congenital NCL, we examined structural changes in brain vessels in CatD-/- mice. Consequently, the mean diameter of the brain vessels in the cerebral cortex on postnatal day 24 (P24) was significantly larger in CatD-/- mice than in wild-type mice. Furthermore, the mean number of brain pericytes in CatD-/- mice began to decline significantly on P16 and almost disappeared on P24, and oxidative DNA damage was first detected in brain pericytes on P12. Examinations with electron microscopy revealed that brain pericytes were laden with dense granular bodies, cytoplasmic vacuoles and lipid droplets. The infiltration of PBMCs characterized by segmented nucleus laden with dense granular bodies was also noted in the cerebral cortex of CatD-/- mice. When primary cultured microglia prepared from enhanced green fluorescent protein (GFP)-expressing transgenic rats were injected into the common carotid artery, GFP-positive microglia were detected in the brain parenchyma of CatD-/-, but not wild-type, mice. Moreover, pepstatin A, a specific aspartic protease inhibitor, induced mitochondria-derived reactive oxygen species (ROS) production in the isolated brain pericytes, which decreased the cell viability. These observations suggest that increased lysosomal storage due to CatD deficiency causes oxidative damage in brain pericytes, subsequently resulting in an increased vessel diameter, enhanced permeability of the BBB and the infiltration of PBMCs. Therefore, protecting brain pericytes against lysosomal storage-induced oxidative stress may represent an alternative treatment strategy for congenital NCL.


Assuntos
Barreira Hematoencefálica/metabolismo , Catepsina D/deficiência , Estresse Oxidativo , Pericitos/metabolismo , Animais , Barreira Hematoencefálica/citologia , Permeabilidade Capilar , Catepsina D/genética , Catepsina D/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pericitos/ultraestrutura
19.
Eur J Neurosci ; 37(5): 816-30, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23279039

RESUMO

Cathepsin C (CC) (EC 3.4.14.1, dipeptidyl peptidase I) is a lysosomal cysteine protease that is required for the activation of several granule-associated serine proteases in vivo. CC has been shown to be constitutively expressed in various tissues, but the enzyme is hardly detectable in central nervous system (CNS) tissues. In the present study, we investigated the regional and cellular distribution of CC in normal, aging and pathological mouse brains. Immunoblotting failed to detect CC protein in whole brain tissues of normal mice, as previously described. However, low proteolytic activity of CC was detected in a brain region-dependent manner, and granular immunohistochemical signals were found in neuronal perikarya of particular brain regions, including the accessory olfactory bulb, the septum, CA2 of the hippocampus, a part of the cerebral cortex, the medial geniculate, and the inferior colliculus. In aged mice, the number of CC-positive neurons increased to some extent. The protein level of CC and its proteolytic activity showed significant increases in particular brain regions of mouse models with pathological conditions--the thalamus in cathepsin D-deficient mice, the hippocampus of ipsilateral brain hemispheres after hypoxic-ischemic brain injury, and peri-damaged portions of brains after penetrating injury. In such pathological conditions, the majority of the cells that were strongly immunopositive for CC were activated microglia. These lines of evidence suggest that CC is involved in normal neuronal function in certain brain regions, and also participates in inflammatory processes accompanying pathogenesis in the CNS.


Assuntos
Lesões Encefálicas/enzimologia , Encéfalo/enzimologia , Catepsina C/metabolismo , Hipóxia-Isquemia Encefálica/enzimologia , Fatores Etários , Animais , Encéfalo/patologia , Catepsina C/genética , Catepsina D/deficiência , Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Neurônios/metabolismo , Especificidade de Órgãos , Proteólise , Regulação para Cima
20.
Neurobiol Dis ; 50: 107-19, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23069676

RESUMO

Cathepsin D deficiency is a fatal neurodegenerative disease characterized by extreme loss of neurons and myelin. Our previous studies have demonstrated that structural and functional alterations in synapses are central to the disease pathogenesis. Therefore, we took a systematic approach to examine the synaptic proteome in cathepsin D knock-out mice, where the synaptic pathology resembles that of human patients. We applied quantitative mass spectrometry analysis on synaptosomal fractions isolated from cathepsin D knock-out and control mice at the age of 24 days. From the approximately 600 identified proteins, 43 were present in different amounts (P<0.05, measured in triple biological replicates) in cathepsin D knock-out mice compared to controls. We connected and bridged these 43 proteins using protein interaction data, and overlaid the network with brain specific gene expression information. Subsequently, we superimposed the network with Gene Ontology, pathway, phenotype and disease involvement, allowing construction of a dynamic, disease-protein centered network and prediction of functional modules. The measured changes in the protein levels, as well as some of the bioinformatically predicted ones, were confirmed by quantitative Western blotting or qualitative immunohistochemistry. This combined approach indicated alterations in distinct cellular entities, previously not associated with the disease, and including microtubule associated cytoskeleton and cell projection organization. Cell spreading and wound healing assays confirmed strongly compromised spatial orientation, associated with changes in distribution of focal adhesions and integrin assembly, in cathepsin D deficient cells. These changes might contribute to commencement of synaptic alterations and neuronal degeneration observed in cathepsin D deficiency.


Assuntos
Encéfalo/metabolismo , Catepsina D/deficiência , Movimento Celular , Citoesqueleto/metabolismo , Animais , Western Blotting , Encéfalo/patologia , Catepsina D/metabolismo , Análise por Conglomerados , Biologia Computacional , Citoesqueleto/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Imunofluorescência , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Lipofuscinoses Ceroides Neuronais/metabolismo , Lipofuscinoses Ceroides Neuronais/patologia , Proteoma , Proteômica , Sinapses
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA