Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
1.
J Clin Invest ; 134(4)2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38357925

RESUMO

NKT cells recognize glycolipids presented by CD1d-expressing antigen-presenting cells (APCs) and include type I NKT cells with antitumor function and type II NKT cells, which have been reported to suppress the antitumor response. Some type II NKT cells recognize sulfatide, a glycosphingolipid with a sulfate modification of the sugar. Type I NKT cells recognize different glycosphingolipids. In this issue of the JCI, Nishio and colleagues showed that APCs could process sulfatide antigens, analogous to protein processing for peptide-reactive T cells. Antigen processing in lysosomes removed sulfate to generate a glycosphingolipid that stimulated type I NKT cells and thereby turned an antigen with no antitumor activity into one that not only stimulated type I NKT cells but also stimulated antitumor responses. These findings may extend to the development of glycolipid antigens that could stimulate anticancer responses via antigen processing by APCs.


Assuntos
Células T Matadoras Naturais , Sulfoglicoesfingolipídeos/metabolismo , Antígenos CD1d , Glicolipídeos/metabolismo , Glicoesfingolipídeos/metabolismo , Sulfatos/metabolismo
2.
J Clin Invest ; 134(4)2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38127463

RESUMO

In a structure-function study of sulfatides that typically stimulate type II NKT cells, we made an unexpected discovery. We compared analogs with sphingosine or phytosphingosine chains and 24-carbon acyl chains with 0-1-2 double bonds (C or pC24:0, 24:1, or 24:2). C24:1 and C24:2 sulfatide presented by the CD1d monomer on plastic stimulated type II, not type I, NKT cell hybridomas, as expected. Unexpectedly, when presented by bone marrow-derived DCs (BMDCs), C24:2 reversed specificity to stimulate type I, not type II, NKT cell hybridomas, mimicking the corresponding ß-galactosylceramide (ßGalCer) without sulfate. C24:2 induced IFN-γ-dependent immunoprotection against CT26 colon cancer lung metastases, skewed the cytokine profile, and activated conventional DC subset 1 cells (cDC1s). This was abrogated by blocking lysosomal processing with bafilomycin A1, or by sulfite blocking of arylsulfatase or deletion of this enyzme that cleaves off sulfate. Thus, C24:2 was unexpectedly processed in BMDCs from a type II to a type I NKT cell-stimulating ligand, promoting tumor immunity. We believe this is the first discovery showing that antigen processing of glycosylceramides alters the specificity for the target cell, reversing the glycolipid's function from stimulating type II NKT cells to stimulating type I NKT cells, thereby introducing protective functional activity in cancer. We also believe our study uncovers a new role for antigen processing that does not involve MHC loading but rather alteration of which type of cell is responding.


Assuntos
Células T Matadoras Naturais , Neoplasias , Humanos , Sulfoglicoesfingolipídeos/metabolismo , Antígenos CD1d/genética , Apresentação de Antígeno , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Sulfatos/metabolismo
3.
Int J Biochem Cell Biol ; 159: 106419, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37086817

RESUMO

Iron chelators, such as deferoxamine, exert an anticancer effect by altering the activity of biomolecules critical for regulation of the cell cycle, cell metabolism, and apoptotic processes. Thus, iron chelators are sometimes used in combination with radio- and/or chemotherapy in the treatment of cancer. The possibility that deferoxamine could induce a program of senescence similar to radio- and/or chemotherapy, fostering adaptation in the treatment of cancer cells, is not fully understood. Using established biochemical techniques, biomarkers linked to lipid composition, and coherent anti-Stokes Raman scattering microscopy, we demonstrated that hepatocellular carcinoma-derived HepG2 cells survive after deferoxamine treatment, acquiring phenotypic traits and representative hallmarks of senescent cells. The results support the view that deferoxamine acts in HepG2 cells to produce oxidative stress-induced senescence by triggering sequential mitochondrial and lysosomal dysfunction accompanied by autophagy blockade. We also focused on the lipidome of senescent cells after deferoxamine treatment. Using mass spectrometry, we found that the deferoxamine-induced senescent cells presented marked remodeling of the phosphoinositol, sulfatide, and cardiolipin profiles, which all play a central role in cell signaling cascades, intracellular membrane trafficking, and mitochondria functions. Detection of alterations in glycosphingolipid sulfate species suggested modifications in ceramide generation, and turnover is frequently described in cancer cell survival and resistance to chemotherapy. Blockade of ceramide generation may explain autophagic default, resistance to apoptosis, and the onset of senescence.


Assuntos
Desferroxamina , Sulfoglicoesfingolipídeos , Humanos , Desferroxamina/farmacologia , Desferroxamina/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Sulfoglicoesfingolipídeos/farmacologia , Células Hep G2 , Quelantes de Ferro/farmacologia , Quelantes de Ferro/metabolismo , Mitocôndrias/metabolismo , Senescência Celular
4.
Neurochem Res ; 48(7): 2059-2065, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36879104

RESUMO

Sulfatides are unique sphingolipids present in the serum and the plasma membrane. Sulfatides exert important functions in a number of systems in the human body, including the nervous, immune, cardiovascular, and coagulation systems.Furthermore, it is closely related to tumor occurrence, development, and metastasis. Peroxisome proliferators-activated receptor α (PPARα) is a class of the nuclear receptor superfamily of transcription factors, which is a potential regulator of sulfatides. This review not only summarizes the current knowledge on the physiological functions of sulfatides in various systems, but also discusses the possible PPARα regulatory mechanisms in sulfatide metabolism and functions. The results of the present analysis provide deep insights and further novel ideas for expanding the research on the physiological function and clinical application of sulfatides.


Assuntos
PPAR alfa , Sulfoglicoesfingolipídeos , Humanos , PPAR alfa/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Fígado/metabolismo , Fatores de Transcrição/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo
5.
Biochim Biophys Acta Gene Regul Mech ; 1865(1): 194777, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34843988

RESUMO

LncRNA (long noncoding RNA) is often dysregulated in tumors especially hepatocellular carcinoma (HCC). However, the dysregulation mechanism of lncRNAs is largely unknown. Here, we showed that lncRNA lncAY expression was stimulated in HCC by either endogenous or exogenous sulfatide. Elevated lncAY promoted HCC cell migration or angiogenesis, whereas lncAY silence suppressed HCC cell migration and proliferation. Interestingly, the activity of lncAY gene promoter was enhanced by sulfatide. Then Myb and MEF2C were identified as the transcription factors responsible for the stimulation of lncAY promoter activity and transcription by sulfatide. Both Myb and MEF2C enrichment on lncAY promoter was further confirmed, and their occupancy on lncAY promoter was strengthened by sulfatide for Myb or MEF2C was acetylated. Mutant Myb-K456A exhibited reduced acetylation and weak stimulation for lncAY transcription. However, Myb mutation K456/503A prevented Myb from acetylation induced by sulfatide. The mutant Myb K456/503A further was unable to occupy lncAY promoter and enhance lncAY transcription. In conclusion, this study demonstrated lncAY transcription was abnormally upregulated by sulfatide in HCC through Myb/MEF2C to promote HCC progression.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Fatores de Transcrição MEF2 , Proteínas Proto-Oncogênicas c-myb , RNA Longo não Codificante , Acetilação , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Fatores de Transcrição MEF2/genética , Proteínas Proto-Oncogênicas c-myb/genética , RNA Longo não Codificante/genética , Sulfoglicoesfingolipídeos/metabolismo
6.
Cells ; 10(12)2021 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-34944026

RESUMO

Sulfatide synthesis in the human renal cancer cell line SMKT-R3 was strongly inhibited in the presence of low µM concentrations of AG-205, a progesterone receptor membrane component 1 (PGRMC1) antagonist. This was also the case in Chinese hamster ovary (CHO) cells stably transfected with UDP-galactose: ceramide galactosyltransferase and cerebroside sulfotransferase, the two enzymes required for sulfatide synthesis. In CHO cells synthesizing galactosylceramide but not sulfatide, galactosylceramide was also strongly reduced, suggesting an effect at the level of galactolipid synthesis. Notably, AG-205 inhibited galactosylceramide synthesis to a similar extent in wild type CHO cells and cells that lack PGRMC1 and/or PGRMC2. In vitro enzyme activity assays showed that AG-205 is an inhibitor of UDP-galactose: ceramide galactosyltransferase, but not cerebroside sulfotransferase. This study shows that PGRMC1 is only one of several targets of AG-205 and should be used with caution, especially in studies using cells synthesizing galactosylceramide and sulfatide.


Assuntos
Inibidores Enzimáticos/farmacologia , Galactosilceramidas/antagonistas & inibidores , Indóis/farmacologia , Neoplasias Renais/tratamento farmacológico , Proteínas de Membrana/genética , Receptores de Progesterona/genética , Sulfoglicoesfingolipídeos/antagonistas & inibidores , Animais , Células CHO , Cricetulus , Galactosilceramidas/biossíntese , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Proteínas de Membrana/antagonistas & inibidores , N-Acilesfingosina Galactosiltransferase , Receptores de Progesterona/antagonistas & inibidores , Sulfoglicoesfingolipídeos/metabolismo , Sulfotransferases/genética , Uridina Difosfato Galactose/genética
7.
Sci Rep ; 11(1): 20513, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34654893

RESUMO

Metachromatic leukodystrophy (MLD) is a lysosomal storage disease caused by an arylsulfatase A (ARSA) deficiency and characterized by severe neurological symptoms resulting from demyelination within the central and peripheral nervous systems. We investigated the feasibility and efficacy of intrathecal administration of a type 9 adeno-associated viral vector encoding ARSA (AAV9/ARSA) for the treatment of 6-week-old MLD model mice, which are presymptomatic, and 1-year-old mice, which exhibit neurological abnormalities. Immunohistochemical analysis following AAV9/ARSA administration showed ARSA expression within the brain, with highest activities in the cerebellum and olfactory bulbs. In mice treated at 1 year, alcian blue staining and quantitative analysis revealed significant decreases in stored sulfatide. Behaviorally, mice treated at 1 year showed no improvement in their ability to traverse narrow balance beams as compared to untreated mice. By contrast, MLD mice treated at 6 weeks showed significant decreases in stored sulfatide throughout the entire brain and improved ability to traverse narrow balance beams. These findings suggest intrathecal administration of an AAV9/ARSA vector is a promising approach to treating genetic diseases of the central nervous system, including MLD, though it may be essential to begin therapy before the onset of neurological symptoms.


Assuntos
Cerebrosídeo Sulfatase/genética , Terapia Genética/métodos , Leucodistrofia Metacromática/terapia , Fatores Etários , Animais , Cerebelo/metabolismo , Cerebrosídeo Sulfatase/metabolismo , Dependovirus , Modelos Animais de Doenças , Vetores Genéticos , Injeções Espinhais , Camundongos Knockout , Medula Espinal/metabolismo , Sulfoglicoesfingolipídeos/metabolismo
8.
Mol Neurodegener ; 16(1): 64, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34526055

RESUMO

BACKGROUND: Human genetic association studies point to immune response and lipid metabolism, in addition to amyloid-beta (Aß) and tau, as major pathways in Alzheimer's disease (AD) etiology. Accumulating evidence suggests that chronic neuroinflammation, mainly mediated by microglia and astrocytes, plays a causative role in neurodegeneration in AD. Our group and others have reported early and dramatic losses of brain sulfatide in AD cases and animal models that are mediated by ApoE in an isoform-dependent manner and accelerated by Aß accumulation. To date, it remains unclear if changes in specific brain lipids are sufficient to drive AD-related pathology. METHODS: To study the consequences of CNS sulfatide deficiency and gain insights into the underlying mechanisms, we developed a novel mouse model of adult-onset myelin sulfatide deficiency, i.e., tamoxifen-inducible myelinating glia-specific cerebroside sulfotransferase (CST) conditional knockout mice (CSTfl/fl/Plp1-CreERT), took advantage of constitutive CST knockout mice (CST-/-), and generated CST/ApoE double knockout mice (CST-/-/ApoE-/-), and assessed these mice using a broad range of methodologies including lipidomics, RNA profiling, behavioral testing, PLX3397-mediated microglia depletion, mass spectrometry (MS) imaging, immunofluorescence, electron microscopy, and Western blot. RESULTS: We found that mild central nervous system (CNS) sulfatide losses within myelinating cells are sufficient to activate disease-associated microglia and astrocytes, and to increase the expression of AD risk genes (e.g., Apoe, Trem2, Cd33, and Mmp12), as well as previously established causal regulators of the immune/microglia network in late-onset AD (e.g., Tyrobp, Dock, and Fcerg1), leading to chronic AD-like neuroinflammation and mild cognitive impairment. Notably, neuroinflammation and mild cognitive impairment showed gender differences, being more pronounced in females than males. Subsequent mechanistic studies demonstrated that although CNS sulfatide losses led to ApoE upregulation, genetically-induced myelin sulfatide deficiency led to neuroinflammation independently of ApoE. These results, together with our previous studies (sulfatide deficiency in the context of AD is mediated by ApoE and accelerated by Aß accumulation) placed both Aß and ApoE upstream of sulfatide deficiency-induced neuroinflammation, and suggested a positive feedback loop where sulfatide losses may be amplified by increased ApoE expression. We also demonstrated that CNS sulfatide deficiency-induced astrogliosis and ApoE upregulation are not secondary to microgliosis, and that astrogliosis and microgliosis seem to be driven by activation of STAT3 and PU.1/Spi1 transcription factors, respectively. CONCLUSION: Our results strongly suggest that sulfatide deficiency is an important contributor and driver of neuroinflammation and mild cognitive impairment in AD pathology.


Assuntos
Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Transtornos da Memória/metabolismo , Bainha de Mielina/química , Doenças Neuroinflamatórias/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Idade de Início , Doença de Alzheimer/etiologia , Aminopiridinas/toxicidade , Animais , Apolipoproteínas E/metabolismo , Química Encefálica , Sistema Nervoso Central/metabolismo , Disfunção Cognitiva/etiologia , Perfilação da Expressão Gênica , Gliose/metabolismo , Humanos , Transtornos da Memória/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Knockout para ApoE , Teste do Labirinto Aquático de Morris , Neuroglia/enzimologia , Neuroglia/fisiologia , Doenças Neuroinflamatórias/etiologia , Teste de Campo Aberto , Proteínas Proto-Oncogênicas/fisiologia , Pirróis/toxicidade , Fator de Transcrição STAT3/fisiologia , Sulfoglicoesfingolipídeos/análise , Sulfotransferases/deficiência , Transativadores/fisiologia
9.
J Biol Chem ; 297(3): 101064, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34375644

RESUMO

An inherited deficiency of arylsulfatase A (ASA) causes the lysosomal storage disease metachromatic leukodystrophy (MLD) characterized by massive intralysosomal storage of the acidic glycosphingolipid sulfatide and progressive demyelination. Lyso-sulfatide, which differs from sulfatide by the lack of the N-linked fatty acid, also accumulates in MLD and is considered a key driver of pathology although its concentrations are far below sulfatide levels. However, the metabolic origin of lyso-sulfatide is unknown. We show here that ASA-deficient murine macrophages and microglial cells express an endo-N-deacylase that cleaves the N-linked fatty acid from sulfatide. An ASA-deficient astrocytoma cell line devoid of this activity was used to identify the enzyme by overexpressing 13 deacylases with potentially matching substrate specificities. Hydrolysis of sulfatide was detected only in cells overexpressing the enzyme fatty acid amide hydrolase (FAAH). A cell-free assay with recombinant FAAH confirmed the novel role of this enzyme in sulfatide hydrolysis. Consistent with the in vitro data, deletion of FAAH lowered lyso-sulfatide levels in a mouse model of MLD. Regardless of the established cytotoxicity of lyso-sulfatide and the anti-inflammatory effects of FAAH inhibition seen in mouse models of several neurological diseases, genetic inactivation of FAAH did not mitigate, but rather exacerbated the disease phenotype of MLD mice. This unexpected finding was reflected by worsening of rotarod performance, increase of anxiety-related exploratory activity, aggravation of peripheral neuropathy, and reduced life expectancy. Thus, we conclude that FAAH has a protective function in MLD and may represent a novel therapeutic target for treatment of this fatal condition.


Assuntos
Amidoidrolases/metabolismo , Leucodistrofia Metacromática/patologia , Psicosina/análogos & derivados , Amidoidrolases/genética , Amidoidrolases/fisiologia , Animais , Linhagem Celular , Cerebrosídeo Sulfatase/deficiência , Cerebrosídeo Sulfatase/genética , Modelos Animais de Doenças , Feminino , Leucodistrofia Metacromática/enzimologia , Leucodistrofia Metacromática/genética , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/fisiopatologia , Camundongos , Camundongos Knockout , Microglia/metabolismo , Cultura Primária de Células , Psicosina/genética , Psicosina/metabolismo , Sulfoglicoesfingolipídeos/metabolismo
10.
Mol Cancer Res ; 17(11): 2306-2317, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31427440

RESUMO

Clear cell renal cell carcinoma (ccRCC) is the most common form of kidney cancer and the major cause of mortality for individuals with von Hippel-Lindau (VHL) disease. ccRCC is characterized most frequently by inactivation of VHL tumor suppressor protein that mediates degradation of the alpha subunit of the hypoxia-inducible factor (HIF) transcription factor family. HIF has been implicated in disease progression and the aim of this study was to identify novel HIF target genes that may contribute to ccRCC. We show that GAL3ST1, an enzyme that catalyzes the sulfonation of the plasma membrane sulfolipid sulfatide, is among the top 50 upregulated genes in ccRCC tissue relative to matched normal tissue. Increased expression of GAL3ST1 in primary ccRCC correlates with decreased survival. We show that GAL3ST1 is a HIF target gene whose expression is induced upon VHL loss leading to the accumulation of its enzymatic product sulfatide. Notably, platelets bind more efficiently to renal cancer cells with high GAL3ST1-sulfatide expression than to GAL3ST1-sulfatide-negative counterparts, which protects ccRCC cells against natural killer cell-mediated cytotoxicity. These results suggest that GAL3ST1 is a HIF-responsive gene that may contribute to ccRCC development via promoting cancer cell evasion of immune surveillance. IMPLICATIONS: Cancer development is in part dependent on evasion of immune response. We identify a HIF target gene product GAL3ST1 that may play a role in this critical process.


Assuntos
Carcinoma de Células Renais/genética , Regulação Neoplásica da Expressão Gênica , Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Renais/genética , Sulfoglicoesfingolipídeos/metabolismo , Sulfotransferases/metabolismo , Apoptose , Plaquetas/patologia , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Genes Reporter , Humanos , Hipóxia , Fator 1 Induzível por Hipóxia/genética , Evasão da Resposta Imune , Neoplasias Renais/patologia , Células Matadoras Naturais/patologia , Microscopia de Fluorescência , Modelos Biológicos , Sulfotransferases/genética , Células Tumorais Cultivadas , Regulação para Cima , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
11.
Glycoconj J ; 36(1): 1-11, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30536036

RESUMO

Serum sulfatides are critical glycosphingolipids that are present in lipoproteins and exert anticoagulant effects. A previous study reported decreased levels of serum sulfatides in hemodialysis patients and suggested an association with cardiovascular disease. However, the mechanism of changes in serum sulfatides in chronic kidney dysfunction has not been well investigated. The current study examined whether a chronic kidney disease (CKD) state could decrease serum sulfatide levels using 5/6 nephrectomy (5/6NCKD) mice, an established CKD murine model, and studied the mechanisms contributing to diminished sulfatides. 5/6NCKD mice and sham operation control mice were sacrificed at the 4th or 12th postoperative week (POW) for measurement of serum sulfatide levels. Hepatic sulfatide content, which is the origin of serum sulfatides, and the expression of sulfatide metabolic enzymes in liver tissue were assessed as well. The 5/6NCKD mice developed CKD and showed increased serum creatinine and indoxyl sulfate. The serum levels and hepatic amounts of sulfatides were significantly decreased in 5/6NCKD mice at both 4 and 12 POW, while the degradative enzymes of sulfatides arylsulfatase A and galactosylceramidase were significantly increased. In a Hepa1-6 murine liver cell line, indoxyl sulfate addition caused intracellular levels of sulfatides to decrease and degradative enzymes of sulfatides to increase in a manner comparable to the changes in 5/6NCKD mice liver tissue. In conclusion, chronic kidney dysfunction causes degradation of sulfatides in the liver to decrease serum sulfatide levels. One explanation of these results is that indoxyl sulfate, a uremic toxin, accelerates the degradation of sulfatides in liver tissue.


Assuntos
Insuficiência Renal Crônica/sangue , Sulfoglicoesfingolipídeos/sangue , Animais , Linhagem Celular Tumoral , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Insuficiência Renal Crônica/metabolismo , Sulfoglicoesfingolipídeos/metabolismo
12.
Breast Cancer Res ; 20(1): 133, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30400820

RESUMO

BACKGROUND: We have previously shown that galactosylceramide (GalCer) affects the tumourigenic and metastatic properties of breast cancer cells by acting as an anti-apoptotic molecule. Since GalCer is a precursor molecule in the synthesis of sulfatides, the present study was aimed to define the role of sulfatides in apoptosis and breast cancer progression. METHODS: Expression of GAL3ST1 in breast cancer cell lines and breast cancer tissue specimens was analysed using real-time PCR, western blotting and immunohistochemistry analysis. The amount of sulfatide, GalCer and ceramide was analysed by thin-layer chromatography binding assay and by the modified hydrophilic interaction liquid chromatography coupled with electrospray mass spectrometry methodology. The tumourigenicity of cancer cells was analysed by an in-vivo tumour growth assay. Apoptotic cells were detected based on caspase-3 activation and the TUNEL assay. The interaction of breast cancer cells with P-selectin or E-selectin was analysed using the flow adhesion assay. The ability of sulfatide-expressing cells to activate and aggregate platelets was studied using the flow-cytometry-based aggregation assay. RESULTS: Using two models of breast cancer, T47D cells with blocked synthesis of sulfatide and MDA-MB-231 cells with neosynthesis of this glycosphingolipid, we showed that high sulfatide levels resulted in increased sensitivity of cancer cells to apoptosis induced by hypoxia and doxorubicin in vitro, and decreased their tumourigenicity after transplantation into athymic nu/nu mice. Accordingly, a clinical study on GAL3ST1 expression in invasive ductal carcinoma revealed that its elevated level is associated with better prognosis. Using MDA-MB-231 cells with neosynthesis of sulfatide we also showed that sulfatide is responsible for adhesion of breast cancer cells to P-selectin-expressing cells, including platelets. Sulfatide also acted as an activating molecule, increasing the expression of P-selectin. CONCLUSIONS: This study demonstrates that increased synthesis of sulfatide sensitises cancer cells to microenvironmental stress factors such as hypoxia and anticancer drugs such as doxorubicin. However, sulfatide is probably not directly involved in apoptotic cascades, because its increased synthesis by GAL3ST1 decreased the amounts of its precursor, GalCer, a known anti-apoptotic molecule. On the other hand, our data support the view that sulfatides are malignancy-related adhesive molecules involved in activating and binding P-selectin-expressing platelets to breast cancer cells.


Assuntos
Neoplasias da Mama/patologia , Mama/patologia , Selectina-P/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Sulfurtransferases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Adesão Celular/fisiologia , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Progressão da Doença , Doxorrubicina/farmacologia , Feminino , Galactosilceramidas/metabolismo , Humanos , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Sulfotransferases , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Neuroimmunol ; 323: 28-35, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30196830

RESUMO

Sulfatide is a major glycosphingolipid in myelin and a target for autoantibodies in autoimmune neuropathies. However neuropathy disease models have not been widely established, in part because currently available monoclonal antibodies to sulfatide may not represent the diversity of anti-sulfatide antibody binding patterns found in neuropathy patients. We sought to address this issue by generating and characterising a panel of new anti-sulfatide monoclonal antibodies. These antibodies have sulfatide reactivity distinct from existing antibodies in assays and in binding to peripheral nerve tissues and can be used to provide insights into the pathophysiological roles of anti-sulfatide antibodies in demyelinating neuropathies.


Assuntos
Autoanticorpos/sangue , Membrana Celular/metabolismo , Neuroglia/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Animais , Linhagem Celular Tumoral , Membrana Celular/patologia , Células Cultivadas , Doenças Desmielinizantes/sangue , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Neuroglia/patologia , Ligação Proteica/fisiologia
14.
J Exp Med ; 215(6): 1679-1692, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29728441

RESUMO

Basal-like breast cancer (BLBC) is associated with a poor clinical outcome as a result of the few treatment options and poor therapeutic response. Here, we report that elevated expression of urine diphosphate-galactose ceramide galactosyltransferase (UGT8) specifically occurs in BLBC and predicts poor prognosis in breast cancer patients. UGT8 expression is transcriptionally up-regulated by Sox10, triggering the sulfatide biosynthetic pathway; increased sulfatide activates integrin αVß5-mediated signaling that contributes to BLBC progression. UGT8 expression promotes, whereas UGT8 knockdown suppresses tumorigenicity and metastasis. Importantly, we identify that zoledronic acid (ZA), a marketed drug for treating osteoporosis and bone metastasis, is a direct inhibitor of UGT8, which blocks the sulfatide biosynthetic pathway. Significantly, a clinically achievable dosage of ZA exhibits apparent inhibitory effect on migration, invasion, and lung metastasis of BLBC cells. Together, our study suggests that UGT8 is a potential prognostic indicator and druggable target of BLBC and that pharmacologic inhibition of UGT8 by ZA offers a promising opportunity for treating this challenging disease.


Assuntos
Neoplasias da Mama/patologia , Progressão da Doença , Gangliosídeo Galactosiltransferase/antagonistas & inibidores , Receptores de Vitronectina/metabolismo , Transdução de Sinais , Sulfoglicoesfingolipídeos/metabolismo , Animais , Vias Biossintéticas/efeitos dos fármacos , Neoplasias da Mama/genética , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Feminino , Gangliosídeo Galactosiltransferase/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos SCID , Invasividade Neoplásica , Metástase Neoplásica , Fatores de Transcrição SOXE/metabolismo , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Regulação para Cima/efeitos dos fármacos , Ácido Zoledrônico/farmacologia
15.
Mol Cancer Res ; 16(4): 610-622, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29453316

RESUMO

Integrin αV gene expression is often dysregulated in cancers especially in hepatocellular carcinoma (HCC); however, the mechanism of regulation is poorly understood. Here, it is demonstrated that sulfatide activated integrin αV gene transcription, through histone H3K9/14 acetylation at the promoter, and high integrin αV expression are closely associated with poor prognosis. To elucidate the mechanism of regulation of acetylation, sulfatide-bound proteins were screened by mass spectrometry (MS), and bromodomain containing protein 1 (BRD1) was identified as an interacting protein that also colocalized with sulfatide in HCC cells. BRD1 was also formed a complex with Sp1, which was recruited to the integrin αV gene promoter. Sulfatide was also found to induce BRD1, monocytic leukemia zinc finger (MOZ) and histone acetyltransferase binding to ORC1 (HBO1) acetyltransferase multiprotein complex recruitment to the integrin αV promoter, which is responsible for histone H3K9/14 acetylation. Finally, knockdown of BRD1 limited sulfatide-induced H3K9/14 acetylation and occupancy of MOZ or HBO1 on integrin αV gene promoter.Implications: This study demonstrates that sulfatide interaction with BRD1 mediates acetylation and is important for regulation of integrin αV gene expression. Mol Cancer Res; 16(4); 610-22. ©2018 AACR.


Assuntos
Carcinoma Hepatocelular/metabolismo , Integrina alfaV/genética , Neoplasias Hepáticas/metabolismo , Proteínas Nucleares/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Regulação para Cima , Acetilação , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Histona Acetiltransferases , Chaperonas de Histonas , Humanos , Integrina alfaV/metabolismo , Neoplasias Hepáticas/genética , Masculino , Modelos Moleculares , Metástase Neoplásica , Proteínas Nucleares/química , Prognóstico , Análise de Sobrevida , Análise Serial de Tecidos
16.
Stem Cells Transl Med ; 6(2): 352-368, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28191778

RESUMO

Allogeneic fetal-derived human neural stem cells (hfNSCs) that are under clinical evaluation for several neurodegenerative diseases display a favorable safety profile, but require immunosuppression upon transplantation in patients. Neural progenitors derived from patient-specific induced pluripotent stem cells (iPSCs) may be relevant for autologous ex vivo gene-therapy applications to treat genetic diseases with unmet medical need. In this scenario, obtaining iPSC-derived neural stem cells (NSCs) showing a reliable "NSC signature" is mandatory. Here, we generated human iPSC (hiPSC) clones via reprogramming of skin fibroblasts derived from normal donors and patients affected by metachromatic leukodystrophy (MLD), a fatal neurodegenerative lysosomal storage disease caused by genetic defects of the arylsulfatase A (ARSA) enzyme. We differentiated hiPSCs into NSCs (hiPS-NSCs) sharing molecular, phenotypic, and functional identity with hfNSCs, which we used as a "gold standard" in a side-by-side comparison when validating the phenotype of hiPS-NSCs and predicting their performance after intracerebral transplantation. Using lentiviral vectors, we efficiently transduced MLD hiPSCs, achieving supraphysiological ARSA activity that further increased upon neural differentiation. Intracerebral transplantation of hiPS-NSCs into neonatal and adult immunodeficient MLD mice stably restored ARSA activity in the whole central nervous system. Importantly, we observed a significant decrease of sulfatide storage when ARSA-overexpressing cells were used, with a clear advantage in those mice receiving neonatal as compared with adult intervention. Thus, we generated a renewable source of ARSA-overexpressing iPSC-derived bona fide hNSCs with improved features compared with clinically approved hfNSCs. Patient-specific ARSA-overexpressing hiPS-NSCs may be used in autologous ex vivo gene therapy protocols to provide long-lasting enzymatic supply in MLD-affected brains. Stem Cells Translational Medicine 2017;6:352-368.


Assuntos
Técnicas de Reprogramação Celular , Reprogramação Celular , Cerebrosídeo Sulfatase/biossíntese , Terapia Genética/métodos , Células-Tronco Pluripotentes Induzidas/transplante , Leucodistrofia Metacromática/cirurgia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Linhagem Celular , Movimento Celular , Cerebrosídeo Sulfatase/genética , Técnicas de Cocultura , Modelos Animais de Doenças , Indução Enzimática , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Células-Tronco Pluripotentes Induzidas/enzimologia , Leucodistrofia Metacromática/enzimologia , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/fisiopatologia , Camundongos Endogâmicos NOD , Camundongos SCID , Regeneração Nervosa , Células-Tronco Neurais/enzimologia , Fenótipo , Sulfoglicoesfingolipídeos/metabolismo , Transcriptoma
18.
Glycoconj J ; 33(6): 927-936, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27318478

RESUMO

Sulfatides, a type of glycosphingolipid, are associated with carcinogenesis. Peroxisome proliferator-activated receptor α (PPARα) is involved in the regulation of sulfatide metabolism as well as in cancer development. We previously reported that transgenic (Tg) mice expressing hepatitis C virus core protein (HCVcp) exhibited age-dependent PPARα activation and carcinogenesis in liver. However, the metabolism of sulfatides in hepatocellular carcinoma is unknown. To examine the relationship between sulfatide metabolism, carcinogenesis, HCVcp, and PPARα, age-dependent changes of these factors were examined in HCVcpTg, PPARα inhibitor-treated HCVcpTg, and non-Tg mice. The sulfatide content in liver, the hepatic expression of two key enzymes catalyzing the initial and last reactions in sulfatide synthesis, the hepatic expression of known sulfatide-transferring protein, oxidative stress, and hepatic PPARα expression and its activation were age-dependently increased in HCVcpTg mice. The increased synthesis and accumulation of sulfatides and PPARα activation were significantly enhanced in liver cancer lesions. These changes were attenuated by PPARα inhibitor treatment and not observed in non-Tg mice. These results suggest that HCVcp-induced age-dependent PPARα activation increases synthesis of sulfatides and the resulting sulfatide accumulation affects HCV-related liver cancer. The monitoring of hepatic sulfatide content and the modulation of sulfatide generation by intervention using a PPARα inhibitor might be useful for the prediction and prevention of HCV-related hepatocarcinogenesis, respectively.


Assuntos
Envelhecimento/metabolismo , Hepacivirus/genética , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Proteínas de Neoplasias/metabolismo , PPAR alfa/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Proteínas do Core Viral/biossíntese , Envelhecimento/genética , Envelhecimento/patologia , Animais , Fígado/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , PPAR alfa/genética , Proteínas do Core Viral/genética
19.
Postepy Hig Med Dosw (Online) ; 70: 489-504, 2016 May 09.
Artigo em Polonês | MEDLINE | ID: mdl-27180966

RESUMO

Sulfatides (3-O-sulfogalactosylceramides, sulfated galactocerebrosides, SM4) are esters of sulfuric acid with galactosylceramides. These acidic glycosphingolipids, present at the external leaflet of the plasma membrane, are synthesized by a variety of mammalian cells. They are especially abundant in the myelin sheath of oligodendrocytes in the central nervous system and Schwann cells in the peripheral nervous system. Studies using cerebroside galactosyltransferase-deficient mice revealed that sulfatides are responsible for proper structure and functioning of myelin. Large amounts of sulfatides are also found in the kidney, gastrointestinal tract, islets of Langerhans, and membranes of erythrocytes, thrombocytes and granulocytes. They are ligands for numerous proteins, but in most cases the biological role of such interactions is poorly understood. A notable exception is their binding by P- and L-selectins. Platelet sulfatides are major ligands for P-selectin, and this interaction is critical for the formation of stable platelet aggregates. Sulfatides also bind to chemokines, and seem to play a role in regulation of cytokine expression in human lymphocytes and monocytes. Aberrant metabolism of sulfatides, could cause several important human diseases. In this article, we describe the changes in sulfatide expression associated with such nervous disorders as metachromatic leukodystrophy (MLD), Parkinson's disease and Alzheimer's disease, and several types of cancer, e.g. colon cancer, kidney cancer, and ovarian cancer. We also discuss the involvement of sulfatides in cancer progression, diabetes and autoimmune and immune disorders such as multiple sclerosis. This acidic glycosphingolipids seem to play an important role in pathogenesis of infectious diseases, serving as receptors for binding various bacteria and viruses.


Assuntos
Sulfoglicoesfingolipídeos/metabolismo , Animais , Doenças Autoimunes/metabolismo , Células Sanguíneas/metabolismo , Membrana Celular/química , Sistema Nervoso Central/metabolismo , Citocinas/metabolismo , Galactosilceramidas/metabolismo , Trato Gastrointestinal/metabolismo , Humanos , Ilhotas Pancreáticas/metabolismo , Rim/metabolismo , Bainha de Mielina/química , Neoplasias/metabolismo , Doenças do Sistema Nervoso/metabolismo , Sistema Nervoso Periférico/metabolismo , Selectinas/metabolismo
20.
Oncotarget ; 7(24): 36563-36576, 2016 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-27145276

RESUMO

Integrin αVß3 is a malignant driver of anchorage-independence and tumor angiogenesis, but its dysregulation in hepatocellular carcinoma (HCC) remains unclear. In this study, we observed that sulfatide significantly promoted integrin αV(ITGAV) expression and wound closure in HCC. We also noted that elevated sulfatide profoundly stimulated integrin αVß3 clustering and signaling. In the cells with integrin αVß3 clustering induced by sulfatide, integrin ß3 subunit was phosphorylated. Simultaneously, focal adhesion kinase (FAK), Src and paxillin were also phosphorylated. Treatment with FAK inhibitor resulted in robust suppression of FAK-Y397 and Src-Y416 phosphorylation stimulated by sulfatide, but not suppression of integrin ß3 phosphorylation. Src inhibitors repressed Src-Y416 and FAK Y861 and Y925 phosphorylation, but not FAK-Y397 and integrin ß3 phosphorylation. After mutation of integrin ß3 (Y773F and Y785F), FAK or Src phosphorylation failed to be stimulated by sulfatide. Moreover, ß3 Y773 and Y785 phosphorylation was suppressed by insulin-like growth factor receptor knockdown even in cells stimulated by sulfatide. In assays of immunoprecipitation and immunostaining with integrin αV or ß3 antibody, labeled sulfatide was found in the complex and co-localized with integrin αVß3. Taken together, this study demonstrated that elevated sulfatide bound to integrin αVß3 and induced clustering and phosphorylation of αVß3 instead of matrix ligand binding, triggering outside-in signaling.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Integrina alfaVbeta3/genética , Transdução de Sinais/efeitos dos fármacos , Sulfoglicoesfingolipídeos/farmacologia , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Paxilina/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Sulfoglicoesfingolipídeos/metabolismo , Tirosina/genética , Tirosina/metabolismo , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA