Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Hum Mol Genet ; 32(7): 1090-1101, 2023 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-36300303

RESUMEN

Cysteamine is currently the only therapy for nephropathic cystinosis. It significantly improves life expectancy and delays progression to end-stage kidney disease; however, it cannot prevent it. Unfortunately, compliance to therapy is often weak, particularly during adolescence. Therefore, finding better treatments is a priority in the field of cystinosis. Previously, we found that genistein, an isoflavone particularly enriched in soy, can revert part of the cystinotic cellular phenotype that is not sensitive to cysteamine in vitro. To test the effects of genistein in vivo, we fed 2-month-old wild-type and Ctns-/- female mice with either a control diet, a genistein-containing diet or a cysteamine-containing diet for 14 months. Genistein (160 mg/kg/day) did not affect the growth of the mice or hepatic functionality. Compared with untreated mice at 16 months, Ctns-/- mice fed with genistein had lower cystine concentrations in their kidneys, reduced formation of cystine crystals, a smaller number of LAMP1-positive structures and an overall better-preserved parenchymal architecture. Cysteamine (400 mg/kg/day) was efficient in reverting the lysosomal phenotype and in preventing the development of renal lesions. These preclinical data indicate that genistein ameliorates kidney injury resulting from cystinosis with no side effects. Genistein therapy represents a potential treatment to improve the outcome for patients with cystinosis.


Asunto(s)
Cistinosis , Enfermedades Renales , Animales , Femenino , Ratones , Cisteamina/uso terapéutico , Cistina/uso terapéutico , Cistinosis/tratamiento farmacológico , Cistinosis/genética , Modelos Animales de Enfermedad , Genisteína/farmacología , Genisteína/uso terapéutico , Riñón
2.
Hum Mol Genet ; 31(13): 2262-2278, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35137071

RESUMEN

Recessive mutations in the CTNS gene encoding the lysosomal transporter cystinosin cause cystinosis, a lysosomal storage disease leading to kidney failure and multisystem manifestations. A Ctns knockout mouse model recapitulates features of cystinosis, but the delayed onset of kidney manifestations, phenotype variability and strain effects limit its use for mechanistic and drug development studies. To provide a better model for cystinosis, we generated a Ctns knockout rat model using CRISPR/Cas9 technology. The Ctns-/- rats display progressive cystine accumulation and crystal formation in multiple tissues including kidney, liver and thyroid. They show an early onset and progressive loss of urinary solutes, indicating generalized proximal tubule dysfunction, with development of typical swan-neck lesions, tubulointerstitial fibrosis and kidney failure, and decreased survival. The Ctns-/- rats also present crystals in the cornea, and bone and liver defects, as observed in patients. Mechanistically, the loss of cystinosin induces a phenotype switch associating abnormal proliferation and dedifferentiation, loss of apical receptors and transporters, and defective lysosomal activity and autophagy in the cells. Primary cultures of proximal tubule cells derived from the Ctns-/- rat kidneys confirmed the key changes caused by cystine overload, including reduced endocytic uptake, increased proliferation and defective lysosomal dynamics and autophagy. The novel Ctns-/- rat model and derived proximal tubule cell system provide invaluable tools to investigate the pathogenesis of cystinosis and to accelerate drug discovery.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros , Cistinosis , Síndrome de Fanconi , Insuficiencia Renal , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Autofagia/genética , Cistina , Cistinosis/genética , Cistinosis/patología , Lisosomas/metabolismo , Ratones , Ratas
3.
J Inherit Metab Dis ; 44(6): 1393-1408, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34494673

RESUMEN

Cystinosis is an inherited metabolic disorder caused by autosomal recessive mutations in the CTNS gene leading to lysosomal cystine accumulation. The disease primarily affects the kidneys followed by extra-renal organ involvement later in life. Azoospermia is one of the unclarified complications which are not improved by cysteamine, which is the only available disease-modifying treatment. We aimed at unraveling the origin of azoospermia in cysteamine-treated cystinosis by confirming or excluding an obstructive factor, and investigating the effect of cysteamine on fertility in the Ctns-/- mouse model compared with wild type. Azoospermia was present in the vast majority of infantile type cystinosis patients. While spermatogenesis was intact, an enlarged caput epididymis and reduced levels of seminal markers for obstruction neutral α-glucosidase (NAG) and extracellular matrix protein 1 (ECM1) pointed towards an epididymal obstruction. Histopathological examination in human and mouse testis revealed a disturbed blood-testis barrier characterized by an altered zonula occludens-1 (ZO-1) protein expression. Animal studies ruled out a negative effect of cysteamine on fertility, but showed that cystine accumulation in the testis is irresponsive to regular cysteamine treatment. We conclude that the azoospermia in infantile cystinosis is due to an obstruction related to epididymal dysfunction, irrespective of the severity of an evolving primary hypogonadism. Regular cysteamine treatment does not affect fertility but has subtherapeutic effects on cystine accumulation in testis.


Asunto(s)
Azoospermia/patología , Barrera Hematotesticular/metabolismo , Cisteamina/uso terapéutico , Cistinosis/tratamiento farmacológico , Testículo/patología , Adulto , Animales , Azoospermia/complicaciones , Azoospermia/genética , Depletores de Cistina/uso terapéutico , Cistinosis/complicaciones , Cistinosis/patología , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Infertilidad Masculina/etiología , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Estudios Retrospectivos , Adulto Joven , Proteína de la Zonula Occludens-1/metabolismo
4.
J Am Soc Nephrol ; 31(7): 1522-1537, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32503896

RESUMEN

BACKGROUND: Mutations in the gene that encodes the lysosomal cystine transporter cystinosin cause the lysosomal storage disease cystinosis. Defective cystine transport leads to intralysosomal accumulation and crystallization of cystine. The most severe phenotype, nephropathic cystinosis, manifests during the first months of life, as renal Fanconi syndrome. The cystine-depleting agent cysteamine significantly delays symptoms, but it cannot prevent progression to ESKD and does not treat Fanconi syndrome. This suggests the involvement of pathways in nephropathic cystinosis that are unrelated to lysosomal cystine accumulation. Recent data indicate that one such potential pathway, lysosome-mediated degradation of autophagy cargoes, is compromised in cystinosis. METHODS: To identify drugs that reduce levels of the autophagy-related protein p62/SQSTM1 in cystinotic proximal tubular epithelial cells, we performed a high-throughput screening on the basis of an in-cell ELISA assay. We then tested a promising candidate in cells derived from patients with, and mouse models of, cystinosis, and in preclinical studies in cystinotic zebrafish. RESULTS: Of 46 compounds identified as reducing p62/SQSTM1 levels in cystinotic cells, we selected luteolin on the basis of its efficacy, safety profile, and similarity to genistein, which we previously showed to ameliorate other lysosomal abnormalities of cystinotic cells. Our data show that luteolin improves the autophagy-lysosome degradative pathway, is a powerful antioxidant, and has antiapoptotic properties. Moreover, luteolin stimulates endocytosis and improves the expression of the endocytic receptor megalin. CONCLUSIONS: Our data show that luteolin improves defective pathways of cystinosis and has a good safety profile, and thus has potential as a treatment for nephropathic cystinosis and other renal lysosomal storage diseases.


Asunto(s)
Antioxidantes/farmacología , Cistinosis/tratamiento farmacológico , Evaluación Preclínica de Medicamentos/métodos , Luteolina/farmacología , ARN Mensajero/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Antioxidantes/efectos adversos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Células Cultivadas , Cistinosis/metabolismo , Modelos Animales de Enfermedad , Endocitosis/efectos de los fármacos , Humanos , Túbulos Renales Proximales/patología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Luteolina/efectos adversos , Lisosomas/efectos de los fármacos , Ratones , Estrés Oxidativo/efectos de los fármacos , Fenotipo , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo , Pez Cebra
5.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34884638

RESUMEN

Diagnosis and cure for rare diseases represent a great challenge for the scientific community who often comes up against the complexity and heterogeneity of clinical picture associated to a high cost and time-consuming drug development processes. Here we show a drug repurposing strategy applied to nephropathic cystinosis, a rare inherited disorder belonging to the lysosomal storage diseases. This approach consists in combining mechanism-based and cell-based screenings, coupled with an affordable computational analysis, which could result very useful to predict therapeutic responses at both molecular and system levels. Then, we identified potential drugs and metabolic pathways relevant for the pathophysiology of nephropathic cystinosis by comparing gene-expression signature of drugs that share common mechanisms of action or that involve similar pathways with the disease gene-expression signature achieved with RNA-seq.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Cistinosis/tratamiento farmacológico , Cistinosis/genética , Reposicionamiento de Medicamentos , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/genética , Enfermedades Raras/tratamiento farmacológico , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/efectos de la radiación , Células Cultivadas , Biología Computacional/métodos , Cistinosis/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Humanos , Enfermedades Renales/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Redes y Vías Metabólicas , Enfermedades Raras/genética , Enfermedades Raras/metabolismo , Transcriptoma
6.
Pediatr Res ; 81(1-1): 113-119, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27656773

RESUMEN

BACKGROUND: Nephropathic cystinosis is a lysosomal storage disease that is caused by mutations in the CTNS gene encoding a cystine/proton symporter cystinosin and an isoform cystinosin-LKG which is generated by an alternative splicing of exon 12. We have investigated the physiological role of the cystinosin-LKG that is widely expressed in epithelial tissues. METHODS: We have analyzed the intracellular localization and the function of the cystinosin-LKG conjugated with DsRed (cystinosin-LKG-RFP) in Madin-Darby canine kidney cells (MDCK II) and in proximal tubular epithelial cells carrying a deletion of the CTNS gene (cystinotic PTEC), respectively. RESULTS: Cystinosin-LKG-RFP colocalized with markers of lysosomes, late endosomes and was also expressed on the apical surface of polarized MDCK II cells. Moreover, immune-electron microscopy images of MDCK II cells overexpressing cystinosin-LKG-RFP showed stacked lamellar membranes inside perinuclear lysosomal structures. To study the role of LKG-isoform, we have investigated cystine accumulation and apoptosis that have been described in cystinotic cells. Cystinosin-LKG decreased cystine levels by approximately 10-fold similarly to cystinosin-RFP. The levels of TNFα- and actinomycin D-inducted apoptosis dropped in cystinotic cells expressing LKG-isoform. This effect was also similar to the main isoform. CONCLUSION: Our results suggest that cystinosin-LKG and cystinosin move similar functional activities in cells.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Cistina/metabolismo , Cistinosis/metabolismo , Cistinosis/patología , Empalme Alternativo , Sistemas de Transporte de Aminoácidos Neutros/química , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Apoptosis , Células Cultivadas , Cistinosis/genética , Perros , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Lisosomas/metabolismo , Células de Riñón Canino Madin Darby , Microscopía Electrónica de Transmisión , Mutación , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
Traffic ; 12(2): 218-31, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21062391

RESUMEN

In this study, we investigated the mechanisms of sterol transport from the plasma membrane (PM) to the endoplasmic reticulum (ER) and lipid droplets (LDs) in HeLa cells. By overexpressing all mammalian oxysterol-binding protein-related proteins (ORPs), we found that especially ORP1S and ORP2 enhanced PM-to-LD sterol transport. This reflected the stimulation of transport from the PM to the ER, rather than from the ER to LDs. Double knockdown of ORP1S and ORP2 inhibited sterol transport from the PM to the ER and LDs, suggesting a physiological role for these ORPs in the process. A two phenylalanines in an acidic tract (FFAT) motif in ORPs that mediates interaction with VAMP-associated proteins (VAPs) in the ER was not necessary for the enhancement of sterol transport by ORPs. However, VAP-A and VAP-B silencing slowed down PM-to-LD sterol transport. This was accompanied by enhanced degradation of ORP2 and decreased levels of several FFAT motif-containing ORPs, suggesting a role for VAPs in sterol transport by stabilization of ORPs.


Asunto(s)
Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Metabolismo de los Lípidos/fisiología , Receptores de Esteroides/metabolismo , Esteroles/metabolismo , Secuencias de Aminoácidos , Transporte Biológico , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Membrana Celular/genética , Colesterol/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Esteroides/genética , Relación Estructura-Actividad , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
8.
Biochim Biophys Acta ; 1821(8): 1089-95, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22266015

RESUMEN

Next to the protein-based machineries composed of small G-proteins, coat complexes, SNAREs and tethering factors, the lipid-based machineries are emerging as important players in membrane trafficking. As a component of these machineries, lipid transfer proteins have recently attracted the attention of cell biologists for their involvement in trafficking along different segments of the secretory pathway. Among these, the four-phosphate adaptor protein 2 (FAPP2) was discovered as a protein that localizes dynamically with the trans-Golgi network and regulates the transport of proteins from the Golgi complex to the cell surface. Later studies have highlighted a role for FAPP2 as lipid transfer protein involved in glycosphingolipid metabolism at the Golgi complex. Here we discuss the available evidence on the function of FAPP2 in both membrane trafficking and lipid metabolism and propose a mechanism of action of FAPP2 that integrates its activities in membrane trafficking and in lipid transfer. This article is part of a Special Issue entitled Lipids and Vesicular Transport.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Glicoesfingolípidos/metabolismo , Lípidos/biosíntesis , Vesículas Transportadoras/metabolismo , Red trans-Golgi/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Animales , Transporte Biológico , Membrana Celular/metabolismo , Expresión Génica , Humanos , Metabolismo de los Lípidos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia
9.
Cells ; 11(3)2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35159136

RESUMEN

Epithelial cells that form the kidney proximal tubule (PT) rely on an intertwined ecosystem of vesicular membrane trafficking pathways to ensure the reabsorption of essential nutrients-a key requisite for homeostasis. The endolysosome stands at the crossroads of this sophisticated network, internalizing molecules through endocytosis, sorting receptors and nutrient transporters, maintaining cellular quality control via autophagy, and toggling the balance between PT differentiation and cell proliferation. Dysregulation of such endolysosome-guided trafficking pathways might thus lead to a generalized dysfunction of PT cells, often causing chronic kidney disease and life-threatening complications. In this review, we highlight the biological functions of endolysosome-residing proteins from the perspectives of understanding-and potentially reversing-the pathophysiology of rare inherited diseases affecting the kidney PT. Using cystinosis as a paradigm of endolysosome disease causing PT dysfunction, we discuss how the endolysosome governs the homeostasis of specialized epithelial cells. This review also provides a critical analysis of the molecular mechanisms through which defects in autophagy pathways can contribute to PT dysfunction, and proposes potential interventions for affected tissues. These insights might ultimately accelerate the discovery and development of new therapeutics, not only for cystinosis, but also for other currently intractable endolysosome-related diseases, eventually transforming our ability to regulate homeostasis and health.


Asunto(s)
Cistinosis , Enfermedades Metabólicas , Autofagia , Cistinosis/metabolismo , Ecosistema , Endosomas/metabolismo , Humanos , Lisosomas/metabolismo , Enfermedades Metabólicas/metabolismo
10.
EMBO Mol Med ; 13(7): e13067, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34165243

RESUMEN

Nephropathic cystinosis is a severe monogenic kidney disorder caused by mutations in CTNS, encoding the lysosomal transporter cystinosin, resulting in lysosomal cystine accumulation. The sole treatment, cysteamine, slows down the disease progression, but does not correct the established renal proximal tubulopathy. Here, we developed a new therapeutic strategy by applying omics to expand our knowledge on the complexity of the disease and prioritize drug targets in cystinosis. We identified alpha-ketoglutarate as a potential metabolite to bridge cystinosin loss to autophagy, apoptosis and kidney proximal tubule impairment in cystinosis. This insight combined with a drug screen revealed a bicalutamide-cysteamine combination treatment as a novel dual-target pharmacological approach for the phenotypical correction of cystinotic kidney proximal tubule cells, patient-derived kidney tubuloids and cystinotic zebrafish.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros , Cistinosis , Sistemas de Transporte de Aminoácidos Neutros/genética , Anilidas , Animales , Cisteamina , Cistinosis/tratamiento farmacológico , Humanos , Nitrilos , Fenotipo , Compuestos de Tosilo , Pez Cebra
11.
J Cell Biol ; 218(3): 783-797, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30659099

RESUMEN

Phosphatidylinositol-4-phosphate (PI4P), a phosphoinositide with key roles in the Golgi complex, is made by Golgi-associated phosphatidylinositol-4 kinases and consumed by the 4-phosphatase Sac1 that, instead, is an ER membrane protein. Here, we show that the contact sites between the ER and the TGN (ERTGoCS) provide a spatial setting suitable for Sac1 to dephosphorylate PI4P at the TGN. The ERTGoCS, though necessary, are not sufficient for the phosphatase activity of Sac1 on TGN PI4P, since this needs the phosphatidyl-four-phosphate-adaptor-protein-1 (FAPP1). FAPP1 localizes at ERTGoCS, interacts with Sac1, and promotes its in-trans phosphatase activity in vitro. We envision that FAPP1, acting as a PI4P detector and adaptor, positions Sac1 close to TGN domains with elevated PI4P concentrations allowing PI4P consumption. Indeed, FAPP1 depletion induces an increase in TGN PI4P that leads to increased secretion of selected cargoes (e.g., ApoB100), indicating that FAPP1, by controlling PI4P levels, acts as a gatekeeper of Golgi exit.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Retículo Endoplásmico/genética , Aparato de Golgi/genética , Células HeLa , Células Hep G2 , Humanos , Proteínas de la Membrana/genética , Ratones , Fosfatos de Fosfatidilinositol/genética
12.
J Cell Biol ; 218(3): 1055-1065, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30659100

RESUMEN

ER-TGN contact sites (ERTGoCS) have been visualized by electron microscopy, but their location in the crowded perinuclear area has hampered their analysis via optical microscopy as well as their mechanistic study. To overcome these limits we developed a FRET-based approach and screened several candidates to search for molecular determinants of the ERTGoCS. These included the ER membrane proteins VAPA and VAPB and lipid transfer proteins possessing dual (ER and TGN) targeting motifs that have been hypothesized to contribute to the maintenance of ERTGoCS, such as the ceramide transfer protein CERT and several members of the oxysterol binding proteins. We found that VAP proteins, OSBP1, ORP9, and ORP10 are required, with OSBP1 playing a redundant role with ORP9, which does not involve its lipid transfer activity, and ORP10 being required due to its ability to transfer phosphatidylserine to the TGN. Our results indicate that both structural tethers and a proper lipid composition are needed for ERTGoCS integrity.


Asunto(s)
Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Lípidos de la Membrana/metabolismo , Receptores de Esteroides/metabolismo , Secuencias de Aminoácidos , Transporte Biológico Activo/fisiología , Retículo Endoplásmico/genética , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/genética , Aparato de Golgi/ultraestructura , Células HeLa , Humanos , Lípidos de la Membrana/genética , Microscopía Electrónica , Receptores de Esteroides/genética
13.
PLoS One ; 11(5): e0154805, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27148969

RESUMEN

Cystinosin mediates an ATP-dependent cystine efflux from lysosomes and causes, if mutated, nephropathic cystinosis, a rare inherited lysosomal storage disease. Alternative splicing of the last exon of the cystinosin sequence produces the cystinosin-LKG isoform that is characterized by a different C-terminal region causing changes in the subcellular distribution of the protein. We have constructed RFP-tagged proteins and demonstrated by site-directed mutagenesis that the carboxyl-terminal SSLKG sequence of cystinosin-LKG is an important sorting motif that is required for efficient targeting the protein to the plasma membrane, where it can mediate H+ coupled cystine transport. Deletion of the SSLKG sequence reduced cystinosin-LKG expression in the plasma membrane and cystine transport by approximately 30%, and induced significant accumulation of the protein in the Golgi apparatus and in lysosomes. Cystinosin-LKG, unlike the canonical isoform, also moves to the lysosomes by the indirect pathway, after endocytic retrieval from the plasma membrane, mainly by a clathrin-mediated endocytosis. Nevertheless, silencing of AP-2 triggers the clathrin-independent endocytosis, showing the complex adaptability of cystinosin-LKG trafficking.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Isoformas de Proteínas/metabolismo , Secuencias de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/química , Membrana Celular/metabolismo , Humanos , Isoformas de Proteínas/química
14.
Curr Opin Cell Biol ; 35: 43-50, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25950841

RESUMEN

Although they were identified as long ago as the 1960s, there are still many unknowns regarding the functions and composition of membrane contact sites between the endoplasmic reticulum (ER) and the trans-Golgi (TG). While it seems to be fairly well established that they facilitate lipid exchange between the two organelles, much less is known about how they are regulated. A bottleneck in the study of the ER-TG contact sites has been the absence of methods for their biochemical isolation and visualization by light microscopy. Herein we provide an overview of current knowledge about ER-TG contact sites with a particular emphasis on the questions that remain to be explored.


Asunto(s)
Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Animales , Transporte Biológico , Membranas Intracelulares/metabolismo , Metabolismo de los Lípidos , Lípidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA