Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Nature ; 623(7989): 1062-1069, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37968398

RESUMO

Endomembrane damage represents a form of stress that is detrimental for eukaryotic cells1,2. To cope with this threat, cells possess mechanisms that repair the damage and restore cellular homeostasis3-7. Endomembrane damage also results in organelle instability and the mechanisms by which cells stabilize damaged endomembranes to enable membrane repair remains unknown. Here, by combining in vitro and in cellulo studies with computational modelling we uncover a biological function for stress granules whereby these biomolecular condensates form rapidly at endomembrane damage sites and act as a plug that stabilizes the ruptured membrane. Functionally, we demonstrate that stress granule formation and membrane stabilization enable efficient repair of damaged endolysosomes, through both ESCRT (endosomal sorting complex required for transport)-dependent and independent mechanisms. We also show that blocking stress granule formation in human macrophages creates a permissive environment for Mycobacterium tuberculosis, a human pathogen that exploits endomembrane damage to survive within the host.


Assuntos
Endossomos , Membranas Intracelulares , Lisossomos , Macrófagos , Grânulos de Estresse , Humanos , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Endossomos/metabolismo , Endossomos/microbiologia , Endossomos/patologia , Membranas Intracelulares/metabolismo , Membranas Intracelulares/microbiologia , Membranas Intracelulares/patologia , Lisossomos/metabolismo , Lisossomos/microbiologia , Lisossomos/patologia , Mycobacterium tuberculosis/metabolismo , Grânulos de Estresse/metabolismo , Técnicas In Vitro , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia
2.
Rev Physiol Biochem Pharmacol ; 185: 107-152, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-33398504

RESUMO

Being originally discovered as cellular recycling bins, lysosomes are today recognized as versatile signaling organelles that control a wide range of cellular functions that are essential not only for the well-being of normal cells but also for malignant transformation and cancer progression. In addition to their core functions in waste disposal and recycling of macromolecules and energy, lysosomes serve as an indispensable support system for malignant phenotype by promoting cell growth, cytoprotective autophagy, drug resistance, pH homeostasis, invasion, metastasis, and genomic integrity. On the other hand, malignant transformation reduces the stability of lysosomal membranes rendering cancer cells sensitive to lysosome-dependent cell death. Notably, many clinically approved cationic amphiphilic drugs widely used for the treatment of other diseases accumulate in lysosomes, interfere with their cancer-promoting and cancer-supporting functions and destabilize their membranes thereby opening intriguing possibilities for cancer therapy. Here, we review the emerging evidence that supports the supplementation of current cancer therapies with lysosome-targeting cationic amphiphilic drugs.


Assuntos
Neoplasias , Humanos , Morte Celular , Neoplasias/metabolismo , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patologia , Lisossomos/metabolismo , Lisossomos/patologia , Transdução de Sinais
3.
J Cell Physiol ; 236(10): 7242-7255, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33843059

RESUMO

Family with sequence similarity 134 member B (FAM134B)/RETREG1/JK1 is a novel gene with recently reported roles in various diseases. Understanding the function and mechanism of action of FAM134B is necessary to develop disease therapies. Notably, emerging data are clarifying the molecular mechanisms of FAM134B function in organelle membrane morphogenesis and the regulation of signaling pathways, such as the Wnt and AKT signaling pathways. In addition, transcription factors, RNA N6 -methyladenosine-mediated epigenetic regulation, microRNA, and small molecules are involved in the regulation of FAM134B expression. This review comprehensively considers recent studies on the role of FAM134B and its potential mechanisms in neurodegenerative diseases, obesity, viral diseases, cancer, and other diseases. The functions of FAM134B in maintaining cell homeostasis by regulating Golgi morphology, endoplasmic reticulum autophagy, and mitophagy are also highlighted, which may be the underlying mechanism of FAM134B gene mutation-induced diseases. Moreover, the molecular mechanisms of the FAM134B function during numerous biological processes are discussed. This review provides novel insights into the functions and mechanisms of FAM134B in various diseases, which will inform the development of effective drugs to treat diseases.


Assuntos
Membranas Intracelulares/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias/metabolismo , Doenças Neurodegenerativas/metabolismo , Obesidade/metabolismo , Organelas/metabolismo , Viroses/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Homeostase , Humanos , Membranas Intracelulares/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Morfogênese , Mutação , Neoplasias/genética , Neoplasias/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Obesidade/genética , Obesidade/patologia , Organelas/genética , Organelas/patologia , Transdução de Sinais , Viroses/genética , Viroses/patologia
4.
Nat Cell Biol ; 23(3): 232-242, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33686253

RESUMO

Lysosomes must maintain the integrity of their limiting membrane to ensure efficient fusion with incoming organelles and degradation of substrates within their lumen. Pancreatic cancer cells upregulate lysosomal biogenesis to enhance nutrient recycling and stress resistance, but it is unknown whether dedicated programmes for maintaining the integrity of the lysosome membrane facilitate pancreatic cancer growth. Using proteomic-based organelle profiling, we identify the Ferlin family plasma membrane repair factor Myoferlin as selectively and highly enriched on the membrane of pancreatic cancer lysosomes. Mechanistically, lysosomal localization of Myoferlin is necessary and sufficient for the maintenance of lysosome health and provides an early acting protective system against membrane damage that is independent of the endosomal sorting complex required for transport (ESCRT)-mediated repair network. Myoferlin is upregulated in human pancreatic cancer, predicts poor survival and its ablation severely impairs lysosome function and tumour growth in vivo. Thus, retargeting of plasma membrane repair factors enhances the pro-oncogenic activities of the lysosome.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células , Membranas Intracelulares/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Biomarcadores Tumorais/genética , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Membranas Intracelulares/patologia , Lisossomos/genética , Lisossomos/patologia , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Musculares/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Prognóstico , Transdução de Sinais , Carga Tumoral
5.
Cells ; 10(1)2021 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-33467111

RESUMO

(1) Background: Lipid metabolism is a fundamental hallmark of all tumors, especially of breast cancer. Few studies describe the different lipid metabolisms and sensitivities to the microenvironment of breast cancer cell subtypes that influence the proliferation, aggressiveness, and success of therapy. This study describes the impact of lipid microenvironment on endoplasmic reticulum (ER) membrane and metabolic activity in two breast cancer cell lines with Luminal A and triple-negative breast cancer (TNBC) features. (2) Methods: We investigated the peculiar lipid phenotype of a TNBC cell line, MDA-MB-231, and a Luminal A cell line, MCF7, and their different sensitivity to exogenous fatty acids (i.e., palmitic acid (PA) and docosahexaenoic acid (DHA)). Moreover, we verified the impact of exogenous fatty acids on ER lipid composition. (3) Results: The data obtained demonstrate that MDA-MB-231 cells are more sensitive to the lipid microenvironment and that both PA and DHA are able to remodel their ER membranes with consequences on resident enzyme activity. On the contrary, MCF7 cells are less sensitive to PA, whereas they incorporate DHA, although less efficiently than MDA-MB-231 cells. (4) Conclusions: This study sustains the importance of lipid metabolism as an innovative hallmark to discriminate breast cancer subclasses and to develop personalized and innovative pharmacological strategies. The different sensitivities to the lipid environment shown by MCF7 and MDA-MB-231 cells might be related to cell malignancy and chemoresistance onset. In the future, this new approach could lead to a substantial decrease both in deleterious side effects for the patients and in the cost of entire therapeutic treatments coupled with increased therapy efficiency.


Assuntos
Neoplasias da Mama/metabolismo , Retículo Endoplasmático/metabolismo , Ácidos Graxos , Membranas Intracelulares/metabolismo , Neoplasias da Mama/patologia , Retículo Endoplasmático/patologia , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Feminino , Humanos , Membranas Intracelulares/patologia , Células MCF-7
6.
Nat Commun ; 11(1): 965, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32075961

RESUMO

The sarco-endoplasmic reticulum (SR/ER) plays an important role in the development and progression of many heart diseases. However, many aspects of its structural organization remain largely unknown, particularly in cells with a highly differentiated SR/ER network. Here, we report a cardiac enriched, SR/ER membrane protein, REEP5 that is centrally involved in regulating SR/ER organization and cellular stress responses in cardiac myocytes. In vitro REEP5 depletion in mouse cardiac myocytes results in SR/ER membrane destabilization and luminal vacuolization along with decreased myocyte contractility and disrupted Ca2+ cycling. Further, in vivo CRISPR/Cas9-mediated REEP5 loss-of-function zebrafish mutants show sensitized cardiac dysfunction upon short-term verapamil treatment. Additionally, in vivo adeno-associated viral (AAV9)-induced REEP5 depletion in the mouse demonstrates cardiac dysfunction. These results demonstrate the critical role of REEP5 in SR/ER organization and function as well as normal heart function and development.


Assuntos
Coração/fisiopatologia , Proteínas de Membrana/deficiência , Retículo Sarcoplasmático/patologia , Animais , Cálcio/metabolismo , Células Cultivadas , Estresse do Retículo Endoplasmático , Técnicas de Inativação de Genes , Inativação Gênica , Coração/crescimento & desenvolvimento , Cardiopatias/metabolismo , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo , Peixe-Zebra
7.
EMBO Mol Med ; 11(6)2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31068339

RESUMO

The current clinical care of glioblastomas leaves behind invasive, radio- and chemo-resistant cells. We recently identified mammary-derived growth inhibitor (MDGI/FABP3) as a biomarker for invasive gliomas. Here, we demonstrate a novel function for MDGI in the maintenance of lysosomal membrane integrity, thus rendering invasive glioma cells unexpectedly vulnerable to lysosomal membrane destabilization. MDGI silencing impaired trafficking of polyunsaturated fatty acids into cells resulting in significant alterations in the lipid composition of lysosomal membranes, and subsequent death of the patient-derived glioma cells via lysosomal membrane permeabilization (LMP). In a preclinical model, treatment of glioma-bearing mice with an antihistaminergic LMP-inducing drug efficiently eradicated invasive glioma cells and secondary tumours within the brain. This unexpected fragility of the aggressive infiltrating cells to LMP provides new opportunities for clinical interventions, such as re-positioning of an established antihistamine drug, to eradicate the inoperable, invasive, and chemo-resistant glioma cells from sustaining disease progression and recurrence.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteína 3 Ligante de Ácido Graxo/metabolismo , Glioblastoma , Membranas Intracelulares , Lisossomos , Proteínas de Neoplasias/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Xenoenxertos , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patologia , Lisossomos/metabolismo , Lisossomos/patologia , Masculino , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Permeabilidade , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Sci Rep ; 9(1): 3265, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30824759

RESUMO

c-Src is a membrane-associated tyrosine kinase that has key roles in the signaling transduction that controls cell growth, adhesion, and migration. In the early stage of carcinogenesis, c-Src is activated under the plasma membrane and transduces oncogenic signals. Here we show that c-Src localized to the endosomal membrane has unique functions in c-Src-transformed cells. Our results indicate that activated c-Src in the endosomal membrane promoted the secretion of exosomes, in which c-Src was encapsulated. In addition, the ESCRT-interacting molecule, Alix was identified as a c-Src-interacting protein in exosomes. We revealed that the interaction between the SH3 domain of c-Src and the proline-rich region of Alix activates ESCRT-mediated intra-luminal vesicle (ILV) formation, resulting in the upregulation of exosome secretion in c-Src-transformed cells. We observed also a correlation between malignant phenotypes and Alix-dependent aberrant exosome secretion in Src-upregulated cancer cells. Collectively, our findings provide a unique mechanism for the upregulation of exosomes in cancer cells, as well as new insights into the significance of exosome secretion in cancer progression.


Assuntos
Proteína Tirosina Quinase CSK/metabolismo , Exossomos/enzimologia , Membranas Intracelulares/enzimologia , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimologia , Proteína Tirosina Quinase CSK/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Exossomos/genética , Exossomos/patologia , Células HCT116 , Células HT29 , Humanos , Membranas Intracelulares/patologia , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Domínios de Homologia de src
9.
PLoS One ; 13(9): e0204532, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30240452

RESUMO

PIKfyve, an evolutionarily conserved kinase synthesizing PtdIns5P and PtdIns(3,5)P2, is crucial for mammalian cell proliferation and viability. Accordingly, PIKfyve inhibitors are now in clinical trials as anti-cancer drugs. Among those, apilimod is the most promising, yet its potency to inhibit PIKfyve and affect endomembrane homeostasis is only partially characterized. We demonstrate here for the first time that apilimod powerfully inhibited in vitro synthesis of PtdIns5P along with that of PtdIns(3,5)P2. HPLC-based resolution of intracellular phosphoinositides (PIs) revealed that apilimod triggered a marked reduction of both lipids in the context of intact cells. Notably, there was also a profound rise in PtdIns3P resulting from arrested PtdIns3P consumption for PtdIns(3,5)P2 synthesis. As typical for PIKfyve inhibition and the concomitant PtdIns(3,5)P2 reduction, apilimod induced the appearance of dilated endomembrane structures in the form of large translucent cytoplasmic vacuoles. Remarkably, bafilomycin A1 (BafA1) fully reversed the aberrant cell phenotype back to normal and completely precluded the appearance of cytoplasmic vacuoles when added prior to apilimod. Inspection of the PI profiles ruled out restoration of the reduced PtdIns(3,5)P2 pool as a molecular mechanism underlying BafA1 rescue. Rather, we found that BafA1 markedly attenuated the PtdIns3P elevation under PIKfyve inhibition. This was accompanied by profoundly decreased endosomal recruitment of fusogenic EEA1. Together, our data demonstrate that apilimod inhibits not only PtdIns(3,5)P2 but also PtdIns5P synthesis and that the cytoplasmic vacuolization triggered by the inhibitor is precluded or reversed by BafA1 through a mechanism associated, in part, with reduction in both PtdIns3P levels and EEA1 membrane recruitment.


Assuntos
Antineoplásicos/farmacologia , Endossomos/efeitos dos fármacos , Membranas Intracelulares/efeitos dos fármacos , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Triazinas/farmacologia , Animais , Células COS , Chlorocebus aethiops , Citoplasma/efeitos dos fármacos , Citoplasma/patologia , Citoplasma/fisiologia , Endossomos/patologia , Endossomos/fisiologia , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , Hidrazonas , Membranas Intracelulares/patologia , Membranas Intracelulares/fisiologia , Macrolídeos/farmacologia , Pirimidinas
10.
Cell Death Dis ; 9(9): 904, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30185776

RESUMO

The molecular mechanisms underlying the severe lung pathology that occurs during SARS-CoV infections remain incompletely understood. The largest of the SARS-CoV accessory protein open reading frames (SARS 3a) oligomerizes, dynamically inserting into late endosomal, lysosomal, and trans-Golgi-network membranes. While previously implicated in a non-inflammatory apoptotic cell death pathway, here we extend the range of SARS 3a pathophysiologic targets by examining its effects on necrotic cell death pathways. We show that SARS 3a interacts with Receptor Interacting Protein 3 (Rip3), which augments the oligomerization of SARS 3a helping drive necrotic cell death. In addition, by inserting into lysosomal membranes SARS 3a triggers lysosomal damage and dysfunction. Consequently, Transcription Factor EB (TFEB) translocates to the nucleus increasing the transcription of autophagy- and lysosome-related genes. Finally, SARS 3a activates caspase-1 either directly or via an enhanced potassium efflux, which triggers NLRP3 inflammasome assembly. In summary, Rip3-mediated oligomerization of SARS 3a causes necrotic cell death, lysosomal damage, and caspase-1 activation-all likely contributing to the clinical manifestations of SARS-CoV infection.


Assuntos
Necrose/virologia , Fases de Leitura Aberta/genética , Síndrome Respiratória Aguda Grave/patologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Células A549 , Apoptose/fisiologia , Autofagia/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Células HeLa , Humanos , Inflamassomos/metabolismo , Membranas Intracelulares/patologia , Membranas Intracelulares/virologia , Lisossomos/metabolismo , Lisossomos/patologia , Lisossomos/virologia , Necrose/metabolismo , Necrose/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Síndrome Respiratória Aguda Grave/virologia
11.
J Cell Biol ; 217(10): 3545-3559, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30006461

RESUMO

Despite their importance as human pathogens, entry of human papillomaviruses (HPVs) into cells is poorly understood. The transmembrane protease γ-secretase executes a crucial function during the early stages of HPV infection, but the role of γ-secretase in infection and the identity of its critical substrate are unknown. Here we demonstrate that γ-secretase harbors a previously uncharacterized chaperone function, promoting low pH-dependent insertion of the HPV L2 capsid protein into endosomal membranes. Upon membrane insertion, L2 recruits the cytosolic retromer, which enables the L2 viral genome complex to enter the retrograde transport pathway and traffic to the Golgi en route for infection. Although a small fraction of membrane-inserted L2 is also cleaved by γ-secretase, this proteolytic event appears dispensable for HPV infection. Our findings demonstrate that γ-secretase is endowed with an activity that can promote membrane insertion of L2, thereby targeting the virus to the productive infectious pathway.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Proteínas do Capsídeo/metabolismo , Papillomavirus Humano 16/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Infecções por Papillomavirus/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Proteínas do Capsídeo/genética , Endossomos/genética , Endossomos/metabolismo , Endossomos/patologia , Endossomos/virologia , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Complexo de Golgi/patologia , Complexo de Golgi/virologia , Células HEK293 , Células HeLa , Papillomavirus Humano 16/genética , Humanos , Concentração de Íons de Hidrogênio , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patologia , Membranas Intracelulares/virologia , Chaperonas Moleculares/genética , Proteínas Oncogênicas Virais/genética , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/patologia , Proteólise
12.
Adv Exp Med Biol ; 997: 225-254, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28815534

RESUMO

Inter-organellar contact sites establish microdomains for localised Ca2+-signalling events. One of these microdomains is established between the ER and the mitochondria. Importantly, the so-called mitochondria-associated ER membranes (MAMs) contain, besides structural proteins and proteins involved in lipid exchange, several Ca2+-transport systems, mediating efficient Ca2+ transfer from the ER to the mitochondria. These Ca2+ signals critically control several mitochondrial functions, thereby impacting cell metabolism, cell death and survival, proliferation and migration. Hence, the MAMs have emerged as critical signalling hubs in physiology, while their dysregulation is an important factor that drives or at least contributes to oncogenesis and tumour progression. In this book chapter, we will provide an overview of the role of the MAMs in cell function and how alterations in the MAM composition contribute to oncogenic features and behaviours.


Assuntos
Sinalização do Cálcio , Retículo Endoplasmático/metabolismo , Membranas Intracelulares/metabolismo , Microdomínios da Membrana/metabolismo , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Animais , Retículo Endoplasmático/patologia , Humanos , Membranas Intracelulares/patologia , Microdomínios da Membrana/patologia , Proteínas de Membrana/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/metabolismo , Neoplasias/patologia , Microambiente Tumoral
13.
Proc Natl Acad Sci U S A ; 114(32): E6507-E6515, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28743750

RESUMO

Serum amyloid A (SAA) is an acute-phase plasma protein that functions in innate immunity and lipid homeostasis. SAA is a protein precursor of reactive AA amyloidosis, the major complication of chronic inflammation and one of the most common human systemic amyloid diseases worldwide. Most circulating SAA is protected from proteolysis and misfolding by binding to plasma high-density lipoproteins. However, unbound soluble SAA is intrinsically disordered and is either rapidly degraded or forms amyloid in a lysosome-initiated process. Although acidic pH promotes amyloid fibril formation by this and many other proteins, the molecular underpinnings are unclear. We used an array of spectroscopic, biochemical, and structural methods to uncover that at pH 3.5-4.5, murine SAA1 forms stable soluble oligomers that are maximally folded at pH 4.3 with ∼35% α-helix and are unusually resistant to proteolysis. In solution, these oligomers neither readily convert into mature fibrils nor bind lipid surfaces via their amphipathic α-helices in a manner typical of apolipoproteins. Rather, these oligomers undergo an α-helix to ß-sheet conversion catalyzed by lipid vesicles and disrupt these vesicles, suggesting a membranolytic potential. Our results provide an explanation for the lysosomal origin of AA amyloidosis. They suggest that high structural stability and resistance to proteolysis of SAA oligomers at pH 3.5-4.5 help them escape lysosomal degradation, promote SAA accumulation in lysosomes, and ultimately damage cellular membranes and liberate intracellular amyloid. We posit that these soluble prefibrillar oligomers provide a missing link in our understanding of the development of AA amyloidosis.


Assuntos
Amiloidose , Membranas Intracelulares , Lisossomos , Multimerização Proteica , Proteína Amiloide A Sérica , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Concentração de Íons de Hidrogênio , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patologia , Lisossomos/química , Lisossomos/metabolismo , Lisossomos/patologia , Camundongos , Estrutura Secundária de Proteína , Proteína Amiloide A Sérica/química , Proteína Amiloide A Sérica/metabolismo
14.
Sci Rep ; 6: 26882, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27246909

RESUMO

Diffuse astrocytoma (DA; WHO grade II) is a low-grade, primary brain neoplasm with high potential of recurrence as higher grade malignant form. We have analyzed differentially expressed membrane proteins from these tumors, using high-resolution mass spectrometry. A total of 2803 proteins were identified, 340 of them differentially expressed with minimum of 2 fold change and based on ≥2 unique peptides. Bioinformatics analysis of this dataset also revealed important molecular networks and pathways relevant to tumorigenesis, mTOR signaling pathway being a major pathway identified. Comparison of 340 differentially expressed proteins with the transcript data from Grade II diffuse astrocytomas reported earlier, revealed about 190 of the proteins correlate in their trends in expression. Considering progressive and recurrent nature of these tumors, we have mapped the differentially expressed proteins for their secretory potential, integrated the resulting list with similar list of proteins from anaplastic astrocytoma (WHO Grade III) tumors and provide a panel of proteins along with their proteotypic peptides, as a resource that would be useful for investigation as circulatory plasma markers for post-treatment surveillance of DA patients.


Assuntos
Astrocitoma/genética , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/genética , Recidiva Local de Neoplasia/genética , Proteoma/genética , Adulto , Sequência de Aminoácidos , Astrocitoma/metabolismo , Astrocitoma/patologia , Astrocitoma/cirurgia , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Estudos de Casos e Controles , Biologia Computacional , Feminino , Perfilação da Expressão Gênica , Humanos , Membranas Intracelulares/química , Membranas Intracelulares/patologia , Masculino , Microssomos/química , Microssomos/patologia , Anotação de Sequência Molecular , Gradação de Tumores , Proteínas de Neoplasias/metabolismo , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/cirurgia , Proteoma/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
15.
Biochim Biophys Acta ; 1863(7 Pt A): 1665-81, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27102538

RESUMO

The potent and selective anti-tumor agent, di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT), localizes in lysosomes and forms cytotoxic copper complexes that generate reactive oxygen species (ROS), resulting in lysosomal membrane permeabilization (LMP) and cell death. Herein, the role of lysosomal membrane stability in the anti-tumor activity of Dp44mT was investigated. Studies were performed using molecules that protect lysosomal membranes against Dp44mT-induced LMP, namely heat shock protein 70 (HSP70) and cholesterol. Up-regulation or silencing of HSP70 expression did not affect Dp44mT-induced LMP in MCF7 cells. In contrast, cholesterol accumulation in lysosomes induced by the well characterized cholesterol transport inhibitor, 3-ß-[2-(diethyl-amino)ethoxy]androst-5-en-17-one (U18666A), inhibited Dp44mT-induced LMP and markedly and significantly (p<0.001) reduced the ability of Dp44mT to inhibit cancer cell proliferation (i.e., increased the IC(50)) by 140-fold. On the other hand, cholesterol extraction using methyl-ß-cyclodextrin enhanced Dp44mT-induced LMP and significantly (p<0.01) increased its anti-proliferative activity. The protective effect of U18666A in increasing lysosomal cholesterol and preventing the cytotoxic activity of Dp44mT was not due to induced autophagy. Instead, U18666A was found to decrease lysosomal turnover, resulting in autophagosome accumulation. Moreover, preincubation with U18666A did not prevent the ability of Dp44mT to induce autophagosome synthesis, indicating that autophagic initiation via Dp44mT occurs independently of LMP. These studies demonstrate the significance of lysosomal membrane stability in relation to the ability of Dp44mT to execute tumor cell death and overcome pro-survival autophagy. Hence, lysosomal-dependent cell death induced by Dp44mT serves as an important anti-tumor strategy. These results are important for comprehensively understanding the mechanism of action of Dp44mT.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Membranas Intracelulares/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Tiossemicarbazonas/farmacologia , Androstenos/farmacologia , Anticolesterolemiantes/farmacologia , Antineoplásicos/metabolismo , Autofagia/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Colesterol/metabolismo , Relação Dose-Resposta a Droga , Feminino , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Concentração Inibidora 50 , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patologia , Lisossomos/metabolismo , Lisossomos/patologia , Células MCF-7 , Permeabilidade , Interferência de RNA , Tiossemicarbazonas/metabolismo , Transfecção , beta-Ciclodextrinas/farmacologia
16.
Biochem Cell Biol ; 92(6): 499-509, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24934166

RESUMO

Protein transport into the human endoplasmic reticulum (ER) is relevant to the biogenesis of most soluble and membrane proteins of organelles, which are involved in endo- or exo-cytsosis. It involves amino-terminal signal peptides in the precursor polypeptides and various transport components in the cytosol plus the ER, and can occur co- or post-translationally. The two mechanisms merge at the level of the ER membrane, specifically at the level of the heterotrimeric Sec61 complex, which forms a dynamic polypeptide-conducting channel in the ER membrane. Since the mammalian ER is also the main intracellular calcium storage organelle, and the Sec61 complex is calcium permeable, the Sec61 complex is tightly regulated in its equilibrium between the closed and open conformations, or "gated", by ligands, such as signal peptides of the transport substrates and the ER lumenal Hsp70-type molecular chaperone BiP. Furthermore, BiP binding to the incoming polypeptide contributes to the efficiency and unidirectionality of transport. Recent insights into the structure and dynamic equilibrium of the Sec61 complex have various mechanistic as well as medical implications.


Assuntos
Canalopatias/metabolismo , Retículo Endoplasmático/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de Membrana/metabolismo , Complexos Multiproteicos/metabolismo , Canalopatias/genética , Canalopatias/patologia , Retículo Endoplasmático/genética , Retículo Endoplasmático/patologia , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Membranas Intracelulares/patologia , Proteínas de Membrana/genética , Complexos Multiproteicos/genética , Sinais Direcionadores de Proteínas , Transporte Proteico/genética , Canais de Translocação SEC
17.
Mol Cell ; 54(1): 133-146, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24703947

RESUMO

Programmed necrotic cell death induced by the tumor necrosis factor alpha (TNF-α) family of cytokines is dependent on a kinase cascade consisting of receptor-interacting kinases RIP1 and RIP3. How these kinase activities cause cells to die by necrosis is not known. The mixed lineage kinase domain-like protein MLKL is a functional RIP3 substrate that binds to RIP3 through its kinase-like domain but lacks kinase activity of its own. RIP3 phosphorylates MLKL at the T357 and S358 sites. Reported here is the development of a monoclonal antibody that specifically recognizes phosphorylated MLKL in cells dying of this pathway and in human liver biopsy samples from patients suffering from drug-induced liver injury. The phosphorylated MLKL forms an oligomer that binds to phosphatidylinositol lipids and cardiolipin. This property allows MLKL to move from the cytosol to the plasma and intracellular membranes, where it directly disrupts membrane integrity, resulting in necrotic death.


Assuntos
Membrana Celular/enzimologia , Proteínas Quinases/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Anticorpos Monoclonais/imunologia , Sítios de Ligação , Cardiolipinas/metabolismo , Membrana Celular/patologia , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Células HT29 , Células HeLa , Humanos , Membranas Intracelulares/enzimologia , Membranas Intracelulares/patologia , Fígado/enzimologia , Fígado/patologia , Lipídeos de Membrana/metabolismo , Necrose , Fosforilação , Conformação Proteica , Proteínas Quinases/genética , Proteínas Quinases/imunologia , Transporte Proteico , Interferência de RNA , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Transdução de Sinais , Especificidade por Substrato , Fatores de Tempo , Transfecção
18.
Biochem Biophys Res Commun ; 443(4): 1232-8, 2014 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-24388981

RESUMO

The endoplasmic reticulum (ER) plays a pivotal role in cellular functions such as the ER stress response. However, the effect of the ER membrane on caspase activation remains unclear. This study reveals that polyglutamine oligomers augmented at ER induce insertion of Bax into the ER membrane, thereby activating caspase-7. In line with the role of ER in cell death induced by polyglutamine expansion, the ER membrane was found to be disrupted and dilated in the brain of a murine model of Huntington's disease. We can conclude that polyglutamine expansion may drive caspase-7 activation by disrupting the ER membrane.


Assuntos
Caspase 7/metabolismo , Retículo Endoplasmático/metabolismo , Doença de Huntington/metabolismo , Peptídeos/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Apoptose , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Retículo Endoplasmático/patologia , Ativação Enzimática , Células HEK293 , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/patologia , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
19.
PLoS Pathog ; 9(6): e1003394, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23762025

RESUMO

Group A Streptococcus (Streptococcus pyogenes or GAS) causes pharyngitis, severe invasive infections, and the post-infectious syndromes of glomerulonephritis and rheumatic fever. GAS can be internalized and killed by epithelial cells in vitro, a process that may contribute to local innate defense against pharyngeal infection. Secretion of the pore-forming toxin streptolysin O (SLO) by GAS has been reported to stimulate targeted autophagy (xenophagy) upon internalization of the bacteria by epithelial cells. Whereas this process was associated with killing of GAS in HeLa cells, studies in human keratinocytes found SLO production enhanced intracellular survival. To reconcile these conflicting observations, we now report in-depth investigation of xenophagy in response to GAS infection of human oropharyngeal keratinocytes, the predominant cell type of the pharyngeal epithelium. We found that SLO expression was associated with prolonged intracellular survival; unexpectedly, expression of the co-toxin NADase was required for this effect. Enhanced intracellular survival was lost upon deletion of NADase or inactivation of its enzymatic activity. Shortly after internalization of GAS by keratinocytes, SLO-mediated damage to the bacteria-containing vacuole resulted in exposure to the cytosol, ubiquitination of GAS and/or associated vacuolar membrane remnants, and engulfment of GAS in LC3-positive vacuoles. We also found that production of streptolysin S could mediate targeting of GAS to autophagosomes in the absence of SLO, a process accompanied by galectin 8 binding to damaged GAS-containing endosomes. Maturation of GAS-containing autophagosome-like vacuoles to degradative autolysosomes was prevented by SLO pore-formation and by SLO-mediated translocation of enzymatically active NADase into the keratinocyte cytosol. We conclude that SLO stimulates xenophagy in pharyngeal keratinocytes, but the coordinated action of SLO and NADase prevent maturation of GAS-containing autophagosomes, thereby prolonging GAS intracellular survival. This novel activity of NADase to block autophagic killing of GAS in pharyngeal cells may contribute to pharyngitis treatment failure, relapse, and chronic carriage.


Assuntos
Autofagia , Células Epiteliais/metabolismo , NAD+ Nucleosidase/metabolismo , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes/metabolismo , Estreptolisinas/biossíntese , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Linhagem Celular Transformada , Sobrevivência Celular , Citosol/metabolismo , Citosol/microbiologia , Citosol/patologia , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patologia , Queratinócitos/metabolismo , Queratinócitos/microbiologia , Queratinócitos/patologia , Viabilidade Microbiana/genética , NAD+ Nucleosidase/genética , Faringite/metabolismo , Faringite/microbiologia , Faringite/patologia , Faringe/metabolismo , Faringe/microbiologia , Faringe/patologia , Infecções Estreptocócicas/genética , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidade , Estreptolisinas/genética , Ubiquitinação , Vacúolos/metabolismo , Vacúolos/microbiologia , Vacúolos/patologia
20.
Mol Pharmacol ; 83(4): 770-80, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23303475

RESUMO

The activity of metabotropic glutamate receptors (mGluRs) is known to be altered as the consequence of neurodegenerative diseases such as Alzheimer, Parkinson, and Huntington disease. However, little attention has been paid to this receptor family's potential link with cancer. Recent reports indicate altered mGluR signaling in various tumor types, and several somatic mutations in mGluR1a in lung cancer were recently described. Group 1 mGluRs (mGluR1a and mGluR5) are coupled primarily to Gαq, leading to the activation of phospholipase C and to the formation of diacylglycerol and inositol 1,4,5-trisphosphate, leading to the release of Ca(2+) from intracellular stores and protein kinase C (PKC) activation. In the present study, we investigated the intracellular localization and G protein-dependent and -independent signaling of eight GRM1 (mGluR1a) somatic mutations. Two mutants found in close proximity to the glutamate binding domain and cysteine-rich region (R375G and G396V) show both decreased cell surface expression and basal inositol phosphate (IP) formation. However, R375G shows increased ERK1/2 activation in response to quisqualate stimulation. A mutant located directly in the glutamate binding site (A168V) shows increased quisqualate-induced IP formation and, similar to R375G, increased ERK1/2 activation. Additionally, a mutation in the G protein-coupled receptor kinase 2/PKC regulatory region (R696W) shows decreased ERK1/2 activation, whereas a mutation within the Homer binding region in the carboxyl-terminal tail (P1148L) does not alter the intracellular localization of the receptor, but it induces changes in cellular morphology and exhibits reduced ERK1/2 activation. Taken together, these results suggest that mGluR1a signaling in cancer is disrupted by somatic mutations with multiple downstream consequences.


Assuntos
Líquido Intracelular/metabolismo , Membranas Intracelulares/metabolismo , Mutação , Neoplasias/genética , Receptores de Glutamato Metabotrópico/genética , Transdução de Sinais/genética , Animais , Regulação para Baixo/genética , Equidae , Variação Genética/genética , Células HEK293 , Humanos , Líquido Intracelular/fisiologia , Membranas Intracelulares/química , Membranas Intracelulares/patologia , Mutação/genética , Neoplasias/metabolismo , Neoplasias/patologia , Receptores de Glutamato Metabotrópico/química , Receptores de Glutamato Metabotrópico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA