Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochem Soc Trans ; 52(1): 407-421, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38348884

RESUMEN

Calcium (Ca2+) regulates a multitude of cellular processes during fertilization and throughout adult life by acting as an intracellular messenger to control effector functions in excitable and non-excitable cells. Changes in intracellular Ca2+ levels are driven by the co-ordinated action of Ca2+ channels, pumps, and exchangers, and the resulting signals are shaped and decoded by Ca2+-binding proteins to drive rapid and long-term cellular processes ranging from neurotransmission and cardiac contraction to gene transcription and cell death. S-acylation, a lipid post-translational modification, is emerging as a critical regulator of several important Ca2+-handling proteins. S-acylation is a reversible and dynamic process involving the attachment of long-chain fatty acids (most commonly palmitate) to cysteine residues of target proteins by a family of 23 proteins acyltransferases (zDHHC, or PATs). S-acylation modifies the conformation of proteins and their interactions with membrane lipids, thereby impacting intra- and intermolecular interactions, protein stability, and subcellular localization. Disruptions of S-acylation can alter Ca2+ signalling and have been implicated in the development of pathologies such as heart disease, neurodegenerative disorders, and cancer. Here, we review the recent literature on the S-acylation of Ca2+ transport proteins of organelles and of the plasma membrane and highlight the molecular basis and functional consequence of their S-acylation as well as the therapeutic potential of targeting this regulation for diseases caused by alterations in cellular Ca2+ fluxes.


Asunto(s)
Proteínas Portadoras , Neoplasias , Humanos , Proteínas Portadoras/metabolismo , Calcio/metabolismo , Ácidos Grasos/metabolismo , Acilación , Aciltransferasas/metabolismo
2.
Cell Rep ; 42(12): 113540, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-38060449

RESUMEN

Store-operated Ca2+ entry (SOCE) mediated by stromal interacting molecule (STIM)-gated ORAI channels at endoplasmic reticulum (ER) and plasma membrane (PM) contact sites maintains adequate levels of Ca2+ within the ER lumen during Ca2+ signaling. Disruption of ER Ca2+ homeostasis activates the unfolded protein response (UPR) to restore proteostasis. Here, we report that the UPR transducer inositol-requiring enzyme 1 (IRE1) interacts with STIM1, promotes ER-PM contact sites, and enhances SOCE. IRE1 deficiency reduces T cell activation and human myoblast differentiation. In turn, STIM1 deficiency reduces IRE1 signaling after store depletion. Using a CaMPARI2-based Ca2+ genome-wide screen, we identify CAMKG2 and slc105a as SOCE enhancers during ER stress. Our findings unveil a direct crosstalk between SOCE and UPR via IRE1, acting as key regulator of ER Ca2+ and proteostasis in T cells and muscles. Under ER stress, this IRE1-STIM1 axis boosts SOCE to preserve immune cell functions, a pathway that could be targeted for cancer immunotherapy.


Asunto(s)
Señalización del Calcio , Calcio , Humanos , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Membrana Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Molécula de Interacción Estromal 1/metabolismo
3.
J Cell Sci ; 136(14)2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37376972

RESUMEN

Changes in membrane phosphoinositides and local Ca2+ elevations at sites of particle capture coordinate the dynamic remodeling of the actin cytoskeleton during phagocytosis. Here, we show that the phosphatidylinositol (PI) transfer proteins PITPNM1 (Nir2) and PITPNM2 (Nir3) maintain phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] homeostasis at phagocytic cups, thereby promoting actin contractility and the sealing of phagosomes. Nir3 and to a lesser extent Nir2 accumulated on endoplasmic reticulum (ER) cisternae juxtaposed to phagocytic cups when expressed in phagocytic COS-7 cells. CRISPR-Cas9 editing of Nir2 and Nir3 genes decreased plasma membrane PI(4,5)P2 levels, store-operated Ca2+ entry (SOCE) and receptor-mediated phagocytosis, stalling particle capture at the cup stage. Re-expression of either Nir2 or Nir3 restored phagocytosis, but not SOCE, proportionally to the PM PI(4,5)P2 levels. Phagosomes forming in Nir2 and Nir3 (Nir2/3) double-knockout cells had decreased overall PI(4,5)P2 levels but normal periphagosomal Ca2+ signals. Nir2/3 depletion reduced the density of contractile actin rings at sites of particle capture, causing repetitive low-intensity contractile events indicative of abortive phagosome closure. We conclude that Nir proteins maintain phosphoinositide homeostasis at phagocytic cups, thereby sustaining the signals that initiate the remodeling of the actin cytoskeleton during phagocytosis.


Asunto(s)
Actinas , Calcio , Actinas/metabolismo , Calcio/metabolismo , Fagocitosis , Membrana Celular/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Fosfatidilinositoles/metabolismo
4.
Contact (Thousand Oaks) ; 6: 25152564221125045, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37366412

RESUMEN

In this news and views, we discuss our recent publication where we described how ER-PM membrane contact sites (MCS) are modulated during store operated calcium entry (SOCE). We also examine why enforcing ER-PM MCS by tethering proteins does not not enhance, but rather inhibits SOCE.

5.
J Cell Sci ; 136(6)2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36807531

RESUMEN

The coronavirus SARS-CoV-2, the agent of the deadly COVID-19 pandemic, is an enveloped virus propagating within the endocytic and secretory organelles of host mammalian cells. Enveloped viruses modify the ionic homeostasis of organelles to render their intra-luminal milieu permissive for viral entry, replication and egress. Here, we show that infection of Vero E6 cells with the delta variant of the SARS-CoV-2 alkalinizes the endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC) as well as lysosomes, mimicking the effect of inhibitors of vacuolar proton ATPases. We further show the envelope protein of SARS-CoV-2 accumulates in the ERGIC when expressed in mammalian cells and selectively dissipates the ERGIC pH. This viroporin action is prevented by mutations of Val25 but not Asn15 within the channel pore of the envelope (E) protein. We conclude that the envelope protein acts as a proton channel in the ERGIC to mitigate the acidity of this intermediate compartment. The altered pH homeostasis of the ERGIC likely contributes to the virus fitness and pathogenicity, making the E channel an attractive drug target for the treatment of COVID-19.


Asunto(s)
COVID-19 , Proteínas del Envoltorio Viral , Animales , Humanos , Proteínas del Envoltorio Viral/metabolismo , Proteínas Viroporinas/metabolismo , COVID-19/metabolismo , Protones , Pandemias , SARS-CoV-2/metabolismo , Aparato de Golgi/metabolismo , Lisosomas/metabolismo , Mamíferos/metabolismo
6.
Cell Calcium ; 105: 102615, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35792400

RESUMEN

Tubular aggregate myopathy (TAM) is a progressive skeletal muscle disease associated with gain-of-function mutations in the ER Ca2+ sensor STIM1 that mediates store-operated Ca2+ entry (SOCE) across the Ca2+-release-activated (CRAC) Ca2+ channel ORAI1. A frameshift mutation in STIM1 inactivation domain, STIM1I484R, was identified in a TAM patient and reported to decrease SOCE. Using ion imaging and electrophysiology, we show that the STIM1I484R mutation instead renders STIM1 constitutively active. In ion imaging experiments, STIM1I484R was less efficient than native STIM1 when expressed alone but enhanced SOCE and increased basal Ca2+ and Mn2+ influx when expressed together with ORAI1. In patch-clamp recordings, STIM1I484R generated larger pre-activated CRAC currents lacking slow Ca2+-dependent inhibition (SCDI). STIM1I484R was pre-recruited in plasma membrane clusters when co-expressed with ORAI1, as were mutants truncated at the frameshift residue or lacking EB-1-binding, which recapitulated STIM1I484R gain-of-function. When expressed alone in human primary myoblasts, STIM1I484R was pre-recruited in large clusters and increased basal Ca2+ entry. These observations establish that STIM1I484R confers a gain of CRAC channel function due to the loss of critical inhibitory C-terminal domains that prevent STIM1 binding to ORAI1, enable STIM1 trapping by microtubules, and mediate SCDI, providing a mechanistic explanation for the muscular defects of TAM patients bearing this mutation.


Asunto(s)
Canales de Calcio , Miopatías Estructurales Congénitas , Calcio/metabolismo , Canales de Calcio/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Microtúbulos/metabolismo , Mutación/genética , Miopatías Estructurales Congénitas/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
7.
Sci Signal ; 15(741): eabm7524, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35857637

RESUMEN

The endoplasmic reticulum (ER) is the largest organelle of the cell and participates in multiple essential functions, including the production of secretory proteins, lipid synthesis, and calcium storage. Sustaining proteostasis requires an intimate coupling with energy production. Mitochondrial respiration evolved to be functionally connected to ER physiology through a physical interface between both organelles known as mitochondria-associated membranes. This quasi-synaptic structure acts as a signaling hub that tunes the function of both organelles in a bidirectional manner and controls proteostasis, cell death pathways, and mitochondrial bioenergetics. Here, we discuss the main signaling mechanisms governing interorganellar communication and their putative role in diseases including cancer and neurodegeneration.


Asunto(s)
Retículo Endoplásmico , Proteostasis , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Metabolismo Energético , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo
8.
J Cell Sci ; 135(6)2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35191477

RESUMEN

Recruitment of STIM proteins to cortical endoplasmic reticulum (cER) domains forming membrane contact sites (MCSs) mediate the store-operated Ca2+ entry (SOCE) pathway essential for human immunity. The cER is dynamically regulated by STIM and tethering proteins during SOCE, but the ultrastructural rearrangement and functional consequences of cER remodeling are unknown. Here, we express natural (E-Syt1 and E-Syt2) and artificial (MAPPER-S and MAPPER-L) protein tethers in HEK-293T cells and correlate the changes in cER length and gap distance, as measured by electron microscopy, with ionic fluxes. We found that native cER cisternae extended during store depletion and remained elongated at a constant ER-plasma membrane (PM) gap distance during subsequent Ca2+ elevations. Tethering proteins enhanced store-dependent cER expansion, anchoring the enlarged cER at tether-specific gap distances of 12-15 nm (E-Syts) and 5-9 nm (MAPPERs). Cells with artificially extended cER had reduced SOCE and reduced agonist-induced Ca2+ release. SOCE remained modulated by calmodulin and exhibited enhanced Ca2+-dependent inhibition. We propose that cER expansion mediated by ER-PM tethering at a close distance negatively regulates SOCE by confining STIM-ORAI complexes to the periphery of enlarged cER sheets, a process that might participate in the termination of store-operated Ca2+ entry.


Asunto(s)
Canales de Calcio , Calcio , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Retículo Endoplásmico/metabolismo , Humanos , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo
9.
Elife ; 102021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34913437

RESUMEN

Efficient immune responses require Ca2+ fluxes across ORAI1 channels during engagement of T cell receptors (TCR) at the immune synapse (IS) between T cells and antigen presenting cells. Here, we show that ZDHHC20-mediated S-acylation of the ORAI1 channel at residue Cys143 promotes TCR recruitment and signaling at the IS. Cys143 mutations reduced ORAI1 currents and store-operated Ca2+ entry in HEK-293 cells and nearly abrogated long-lasting Ca2+ elevations, NFATC1 translocation, and IL-2 secretion evoked by TCR engagement in Jurkat T cells. The acylation-deficient channel remained in cholesterol-poor domains upon enforced ZDHHC20 expression and was recruited less efficiently to the IS along with actin and TCR. Our results establish S-acylation as a critical regulator of ORAI1 channel trafficking and function at the IS and reveal that ORAI1 S-acylation enhances TCR recruitment to the synapse.


Asunto(s)
Aciltransferasas/genética , Calcio/metabolismo , Proteína ORAI1/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Acilación , Aciltransferasas/metabolismo , Células Presentadoras de Antígenos/metabolismo , Células HEK293 , Humanos , Células Jurkat/metabolismo , Microdominios de Membrana/metabolismo , Proteína ORAI1/metabolismo , Azufre/metabolismo , Linfocitos T/metabolismo
10.
Sci Adv ; 7(46): eabe5469, 2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34767445

RESUMEN

Programmed cell death is regulated by the balance between activating and inhibitory signals. Here, we have identified RECS1 (responsive to centrifugal force and shear stress 1) [also known as TMBIM1 (transmembrane BAX inhibitor motif containing 1)] as a proapoptotic member of the TMBIM family. In contrast to other proteins of the TMBIM family, RECS1 expression induces cell death through the canonical mitochondrial apoptosis pathway. Unbiased screening indicated that RECS1 sensitizes cells to lysosomal perturbations. RECS1 localizes to lysosomes, where it regulates their acidification and calcium content, triggering lysosomal membrane permeabilization. Structural modeling and electrophysiological studies indicated that RECS1 is a pH-regulated calcium channel, an activity that is essential to trigger cell death. RECS1 also sensitizes whole animals to stress in vivo in Drosophila melanogaster and zebrafish models. Our results unveil an unanticipated function for RECS1 as a proapoptotic component of the TMBIM family that ignites cell death programs at lysosomes.

12.
Nat Commun ; 11(1): 2401, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32409639

RESUMEN

The molecular connections between homeostatic systems that maintain both genome integrity and proteostasis are poorly understood. Here we identify the selective activation of the unfolded protein response transducer IRE1α under genotoxic stress to modulate repair programs and sustain cell survival. DNA damage engages IRE1α signaling in the absence of an endoplasmic reticulum (ER) stress signature, leading to the exclusive activation of regulated IRE1α-dependent decay (RIDD) without activating its canonical output mediated by the transcription factor XBP1. IRE1α endoribonuclease activity controls the stability of mRNAs involved in the DNA damage response, impacting DNA repair, cell cycle arrest and apoptosis. The activation of the c-Abl kinase by DNA damage triggers the oligomerization of IRE1α to catalyze RIDD. The protective role of IRE1α under genotoxic stress is conserved in fly and mouse. Altogether, our results uncover an important intersection between the molecular pathways that sustain genome stability and proteostasis.


Asunto(s)
Supervivencia Celular/genética , Reparación del ADN , Proteínas de Drosophila/metabolismo , Endorribonucleasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad del ARN/genética , Animales , Daño del ADN , Proteínas de Drosophila/genética , Drosophila melanogaster , Endorribonucleasas/genética , Femenino , Fibroblastos , Inestabilidad Genómica , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Multimerización de Proteína , Proteínas Serina-Treonina Quinasas/genética , Proteostasis/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , ARN Mensajero/metabolismo
14.
Nat Cell Biol ; 21(6): 755-767, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31110288

RESUMEN

Mitochondria-associated membranes (MAMs) are central microdomains that fine-tune bioenergetics by the local transfer of calcium from the endoplasmic reticulum to the mitochondrial matrix. Here, we report an unexpected function of the endoplasmic reticulum stress transducer IRE1α as a structural determinant of MAMs that controls mitochondrial calcium uptake. IRE1α deficiency resulted in marked alterations in mitochondrial physiology and energy metabolism under resting conditions. IRE1α determined the distribution of inositol-1,4,5-trisphosphate receptors at MAMs by operating as a scaffold. Using mutagenesis analysis, we separated the housekeeping activity of IRE1α at MAMs from its canonical role in the unfolded protein response. These observations were validated in vivo in the liver of IRE1α conditional knockout mice, revealing broad implications for cellular metabolism. Our results support an alternative function of IRE1α in orchestrating the communication between the endoplasmic reticulum and mitochondria to sustain bioenergetics.


Asunto(s)
Retículo Endoplásmico/metabolismo , Endorribonucleasas/genética , Metabolismo Energético , Mitocondrias/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Animales , Calcio/metabolismo , Señalización del Calcio/genética , Retículo Endoplásmico/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Ratones , Ratones Noqueados , Mitocondrias/genética
16.
Nat Cell Biol ; 20(8): 942-953, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30013108

RESUMEN

Maintenance of endoplasmic reticulum (ER) proteostasis is controlled by a signalling network known as the unfolded protein response (UPR). Here, we identified filamin A as a major binding partner of the ER stress transducer IRE1α. Filamin A is an actin crosslinking factor involved in cytoskeleton remodelling. We show that IRE1α controls actin cytoskeleton dynamics and affects cell migration upstream of filamin A. The regulation of cytoskeleton dynamics by IRE1α is independent of its canonical role as a UPR mediator, serving instead as a scaffold that recruits and regulates filamin A. Targeting IRE1α expression in mice affected normal brain development, generating a phenotype resembling periventricular heterotopia, a disease linked to the loss of function of filamin A. IRE1α also modulated cell movement and cytoskeleton dynamics in fly and zebrafish models. This study unveils an unanticipated biological function of IRE1α in cell migration, whereby filamin A operates as an interphase between the UPR and the actin cytoskeleton.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Movimiento Celular , Endorribonucleasas/metabolismo , Fibroblastos/metabolismo , Filaminas/metabolismo , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Endorribonucleasas/deficiencia , Endorribonucleasas/genética , Evolución Molecular , Femenino , Filaminas/genética , Células HEK293 , Humanos , Cinética , Masculino , Ratones , Ratones Noqueados , Neuronas/patología , Heterotopia Nodular Periventricular/genética , Heterotopia Nodular Periventricular/metabolismo , Heterotopia Nodular Periventricular/patología , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Respuesta de Proteína Desplegada , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
17.
Mol Cell ; 69(2): 238-252.e7, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29351844

RESUMEN

Maintenance of endoplasmic reticulum (ER) proteostasis is controlled by a dynamic signaling network known as the unfolded protein response (UPR). IRE1α is a major UPR transducer, determining cell fate under ER stress. We used an interactome screening to unveil several regulators of the UPR, highlighting the ER chaperone Hsp47 as the major hit. Cellular and biochemical analysis indicated that Hsp47 instigates IRE1α signaling through a physical interaction. Hsp47 directly binds to the ER luminal domain of IRE1α with high affinity, displacing the negative regulator BiP from the complex to facilitate IRE1α oligomerization. The regulation of IRE1α signaling by Hsp47 is evolutionarily conserved as validated using fly and mouse models of ER stress. Hsp47 deficiency sensitized cells and animals to experimental ER stress, revealing the significance of Hsp47 to global proteostasis maintenance. We conclude that Hsp47 adjusts IRE1α signaling by fine-tuning the threshold to engage an adaptive UPR.


Asunto(s)
Endorribonucleasas/metabolismo , Proteínas del Choque Térmico HSP47/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Proteínas del Choque Térmico HSP47/fisiología , Humanos , Ratones , Chaperonas Moleculares/metabolismo , Transducción de Señal , Estrés Fisiológico , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada
18.
Cell Calcium ; 70: 24-31, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29054537

RESUMEN

Endoplasmic reticulum (ER) calcium signaling is implicated in a myriad of coordinated cellular processes. The ER calcium content is tightly regulated as it allows a favorable environment for protein folding, in addition to operate as a major reservoir for fast and specific release of calcium. Altered ER homeostasis impacts protein folding, activating the unfolded protein response (UPR) as a rescue mechanism to restore proteostasis. ER calcium release impacts mitochondrial metabolism and also fine-tunes the threshold to undergo apoptosis under chronic stress. The global coordination between UPR signaling and energetic demands takes place at mitochondrial associated membranes (MAMs), specialized subdomains mediating interorganelle communication. Here we discuss current models explaining the functional relationship between ER homeostasis and various cellular responses to coordinate proteostasis and metabolic maintenance.


Asunto(s)
Señalización del Calcio , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Animales , Humanos , Modelos Biológicos , Respuesta de Proteína Desplegada
19.
Cell Death Differ ; 24(9): 1478-1487, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28622296

RESUMEN

In the last decade, the endoplasmic reticulum (ER) has emerged as a central organelle regulating the core mitochondrial apoptosis pathway. At the ER membrane, a variety of stress signals are integrated toward determining cell fate, involving a complex cross talk between key homeostatic pathways including the unfolded protein response, autophagy, calcium signaling and mitochondrial bioenergetics. In this context, key regulators of cell death of the BCL-2 and TMBIM/BI-1 family of proteins have relevant functions as stress rheostats mediated by the formation of distinct protein complexes that regulate the switch between adaptive and proapoptotic phases under stress. Here, we overview recent advances on our molecular understanding of how the apoptotic machinery integrates stress signals toward cell fate decisions upstream of the mitochondrial gateway of death.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Autofagia/genética , Autofagia/fisiología , Señalización del Calcio/genética , Señalización del Calcio/fisiología , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/fisiología
20.
Front Oncol ; 7: 55, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28421160

RESUMEN

Endoplasmic reticulum (ER) to mitochondria communication has emerged in recent years as a signaling hub regulating cellular physiology with a relevant contribution to diseases including cancer and neurodegeneration. This functional integration is exerted through discrete interorganelle structures known as mitochondria-associated membranes (MAMs). At these domains, ER/mitochondria physically associate to dynamically adjust metabolic demands and the response to stress stimuli. Here, we provide a focused overview of how the ER shapes the function of the mitochondria, giving a special emphasis to the significance of local signaling of the unfolded protein response at MAMs. The implications to cell fate control and the progression of cancer are also discussed.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...