Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(21): e2318874121, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38753510

RESUMO

The single-pass transmembrane protein Stromal Interaction Molecule 1 (STIM1), located in the endoplasmic reticulum (ER) membrane, possesses two main functions: It senses the ER-Ca2+ concentration and directly binds to the store-operated Ca2+ channel Orai1 for its activation when Ca2+ recedes. At high resting ER-Ca2+ concentration, the ER-luminal STIM1 domain is kept monomeric but undergoes di/multimerization once stores are depleted. Luminal STIM1 multimerization is essential to unleash the STIM C-terminal binding site for Orai1 channels. However, structural basis of the luminal association sites has so far been elusive. Here, we employed molecular dynamics (MD) simulations and identified two essential di/multimerization segments, the α7 and the adjacent region near the α9-helix in the sterile alpha motif (SAM) domain. Based on MD results, we targeted the two STIM1 SAM domains by engineering point mutations. These mutations interfered with higher-order multimerization of ER-luminal fragments in biochemical assays and puncta formation in live-cell experiments upon Ca2+ store depletion. The STIM1 multimerization impeded mutants significantly reduced Ca2+ entry via Orai1, decreasing the Ca2+ oscillation frequency as well as store-operated Ca2+ entry. Combination of the ER-luminal STIM1 multimerization mutations with gain of function mutations and coexpression of Orai1 partially ameliorated functional defects. Our data point to a hydrophobicity-driven binding within the ER-luminal STIM1 multimer that needs to switch between resting monomeric and activated multimeric state. Altogether, these data reveal that interactions between SAM domains of STIM1 monomers are critical for multimerization and activation of the protein.


Assuntos
Cálcio , Retículo Endoplasmático , Simulação de Dinâmica Molecular , Proteínas de Neoplasias , Proteína ORAI1 , Multimerização Proteica , Molécula 1 de Interação Estromal , Molécula 1 de Interação Estromal/metabolismo , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/química , Humanos , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/química , Retículo Endoplasmático/metabolismo , Cálcio/metabolismo , Proteína ORAI1/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/química , Domínios Proteicos , Células HEK293 , Sítios de Ligação , Ligação Proteica
2.
ACS Synth Biol ; 11(8): 2756-2765, 2022 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-35802180

RESUMO

Highly regulated intracellular calcium entry affects numerous cellular physiological events. External regulation of intracellular calcium signaling presents a great opportunity for the artificial regulation of cellular activity. Calcium entry can be mediated by STIM proteins interacting with Orai calcium channels; therefore, the STIM1-Orai1 pair has become a tool for artificially modulating calcium entry. We report on an innovative genetically engineered protease-activated Orai activator called PACE. CAD self-dimerization and activation were inhibited with a coiled-coil forming peptide pair linked to CAD via a protease cleavage site. PACE generated sustained calcium entry after its activation with a reconstituted split protease. We also generated PACE, whose transcriptional activation of NFAT was triggered by PPV or TEV protease. Using PACE, we successfully activated the native NFAT signaling pathway and the production of cytokines in a T-cell line. PACE represents a useful tool for generating sustained calcium entry to initiate calcium-dependent protein translation. PACE provides a promising template for the construction of links between various protease activation pathways and calcium signaling.


Assuntos
Canais de Cálcio , Cálcio , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Proteína ORAI1/química , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Peptídeo Hidrolases/metabolismo
3.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34768945

RESUMO

CRAC, which plays important role in Ca2+-dependent T-lymphocyte activation, is composed of the ER-resident STIM1 and the plasma membrane Orai1 pore-forming subunit. Both accumulate at the immunological synapse (IS) between a T cell and an antigen-presenting cell (APC). We hypothesized that adapter/interacting proteins regulate Orai1 residence in the IS. We could show that mGFP-tagged Orai1-Full channels expressed in Jurkat cells had a biphasic IS-accumulation kinetics peaked at 15 min. To understand the background of Orai1 IS-redistribution we knocked down STIM1 and SAP97 (adaptor protein with a short IS-residency (15 min) and ability to bind Orai1 N-terminus): the mGFP-Orai1-Full channels kept on accumulating in the IS up to the 60th minute in the STIM1- and SAP97-lacking Jurkat cells. Deletion of Orai1 N terminus (mGFP-Orai1-Δ72) resulted in the same time course as described for STIM1/SAP97 knock-down cells. Ca2+-imaging of IS-engaged T-cells revealed that of Orai1 residency modifies the Ca2+-response: cells expressing mGFP-Orai1-Δ72 construct or mGFP-Orai1-Full in SAP-97 knock-down cells showed higher number of Ca2+-oscillation up to the 90th minute after IS formation. Overall, these data suggest that SAP97 may contribute to the short-lived IS-residency of Orai1 and binding of STIM1 to Orai1 N-terminus is necessary for SAP97-Orai1 interaction.


Assuntos
Sinalização do Cálcio/imunologia , Sinapses Imunológicas/metabolismo , Proteína ORAI1/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Imunidade Adaptativa , Proteína 1 Homóloga a Discs-Large/antagonistas & inibidores , Proteína 1 Homóloga a Discs-Large/genética , Proteína 1 Homóloga a Discs-Large/metabolismo , Retículo Endoplasmático/metabolismo , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Sinapses Imunológicas/genética , Sinapses Imunológicas/imunologia , Células Jurkat , Cinética , Ativação Linfocitária , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/química , Proteína ORAI1/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Molécula 1 de Interação Estromal/antagonistas & inibidores , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo
4.
Mol Cell ; 81(23): 4784-4798.e7, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34800360

RESUMO

Calcium influx through plasma membrane calcium release-activated calcium (CRAC) channels, which are formed of hexamers of Orai1, is a potent trigger for many important biological processes, most notably in T cell-mediated immunity. Through a bioinformatics-led cell biological screen, we have identified Orai1 as a substrate for the rhomboid intramembrane protease RHBDL2. We show that RHBDL2 prevents stochastic calcium signaling in unstimulated cells through conformational surveillance and cleavage of inappropriately activated Orai1. A conserved disease-linked proline residue is responsible for RHBDL2's recognizing the active conformation of Orai1, which is required to sharpen switch-like signaling triggered by store-operated calcium entry. Loss of RHBDL2 control of CRAC channel activity causes severe dysregulation of downstream CRAC channel effectors, including transcription factor activation, inflammatory cytokine expression, and T cell activation. We propose that this surveillance function may represent an ancient activity of rhomboid proteases in degrading unwanted signaling proteins.


Assuntos
Proteína ORAI1/química , Peptídeo Hidrolases/química , Serina Endopeptidases/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/química , Sinalização do Cálcio/fisiologia , Membrana Celular/metabolismo , Biologia Computacional , Drosophila melanogaster , Células HEK293 , Humanos , Ativação do Canal Iônico , Ativação Linfocitária , Proteínas de Membrana/metabolismo , Mutação , Ligação Proteica , Conformação Proteica , Transdução de Sinais , Processos Estocásticos
5.
PLoS One ; 16(10): e0258670, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34653219

RESUMO

Molecular steps that activate store-operated calcium entry (SOCE) via Orai channel supramolecular complex remain incompletely defined. We have earlier shown that α-SNAP regulates the on-site functional assembly and calcium selectivity of Orai1 channels. Here we investigate the molecular basis of its association with Orai, Stim and find that the affinity of α-SNAP for Orai and Stim is substantially higher than previously reported affinities between Stim and Orai sub-domains. α-SNAP binds the coiled-coil 3 (CC3) sub-domain of Stim1. Mutations of Tryptophan 430 in Stim1-CC3 disrupted α-SNAP association and SOCE, demonstrating a novel α-SNAP dependent function for this crucial subdomain. Further, α-SNAP binds the hinge region near the C-terminus of Orai1 and an additional broad region near the N-terminus and Valine 262 and Leucine 74 were necessary for these respective interactions, but not Orai, Stim co-clustering. Thus, high affinity interactions with α-SNAP are necessary for imparting functionality to Stim, Orai clusters and induction of SOCE.


Assuntos
Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Clonagem Molecular , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Camundongos , Mutação , Proteínas de Neoplasias/química , Proteína ORAI1/química , Ligação Proteica , Molécula 1 de Interação Estromal/química
6.
Cell Rep ; 35(13): 109322, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34192542

RESUMO

Junctional coupling between endoplasmic reticulum (ER) Ca2+-sensor STIM proteins and plasma membrane (PM) Orai channels mediates Ca2+ signals in most cells. We reveal that PM-tethered, fluorescently tagged C-terminal M4x (fourth transmembrane helix contains a cytoplasmic C-terminal extension) peptides from Orai channels undergo a Leu-specific signature of direct interaction with the STIM1 Orai-activating region (SOAR), exactly mimicking STIM1 binding to gate Orai channels. The 20-amino-acid Orai3-M4x peptide associates avidly with STIM1 within ER-PM junctions, functions to competitively block native Ca2+ signals, and mediates a key modification of STIM-Orai coupling induced by 2-aminoethoxydiphenyl borate. By blocking STIM-Orai coupling, the Orai3-M4x peptide reveals the critical role of Orai channels in driving Ca2+ oscillatory signals and transcriptional control through NFAT. The M4x peptides interact independently with SOAR dimers consistent with unimolecular coupling between Orai subunits and STIM1 dimers. We reveal the critical role of M4x helices in defining the coupling interface between STIM and Orai proteins to mediate store-operated Ca2+ signals.


Assuntos
Sinalização do Cálcio , Proteína ORAI1/química , Proteína ORAI1/metabolismo , Peptídeos/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Compostos de Boro/farmacologia , Canais de Cálcio/metabolismo , Membrana Celular/metabolismo , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Células HEK293 , Células HeLa , Humanos , Ativação do Canal Iônico , Leucina/metabolismo , Modelos Moleculares , Mutação/genética , Fatores de Transcrição NFATC/metabolismo , Ligação Proteica , Multimerização Proteica , Transcrição Gênica/efeitos dos fármacos
7.
J Clin Immunol ; 41(5): 1004-1015, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33650027

RESUMO

Store-operated Ca2+ entry (SOCE) represents a predominant Ca2+ influx pathway in non-excitable cells. SOCE is required for immune cell activation and is mediated by the plasma membrane (PM) channel ORAI1 and the endoplasmic reticulum (ER) Ca2+ sensor STIM1. Mutations in the Orai1 or STIM1 genes abolish SOCE leading to combined immunodeficiency (CID), muscular hypotonia, and anhidrotic ectodermal dysplasia. Here, we identify a novel autosomal recessive mutation in ORAI1 in a child with CID. The patient is homozygous for p.C126R mutation in the second transmembrane domain (TM2) of ORAI1, a region with no previous loss-of-function mutations. SOCE is suppressed in the patient's lymphocytes, which is associated with impaired T cell proliferation and cytokine production. Functional analyses demonstrate that the p.C126R mutation does not alter protein expression but disrupts ORAI1 trafficking. Orai1-C126R does not insert properly into the bilayer resulting in ER retention. Insertion of an Arg on the opposite face of TM2 (L135R) also results in defective folding and trafficking. We conclude that positive side chains within ORAI1 TM2 are not tolerated and result in misfolding, defective bilayer insertion, and channel trafficking thus abolishing SOCE and resulting in CID.


Assuntos
Canalopatias/diagnóstico , Proteína ORAI1/genética , Doenças da Imunodeficiência Primária/diagnóstico , Cálcio/metabolismo , Proliferação de Células , Células Cultivadas , Canalopatias/genética , Canalopatias/imunologia , Citocinas/imunologia , Feminino , Humanos , Lactente , Mutação , Proteína ORAI1/química , Proteína ORAI1/metabolismo , Doenças da Imunodeficiência Primária/genética , Doenças da Imunodeficiência Primária/imunologia , Transporte Proteico , Linfócitos T/imunologia
8.
Cell Calcium ; 94: 102366, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33581587

RESUMO

Store-operated Orai channels are a primary mechanism for mobilizing Ca2+ signals in both non-excitable cells and excitable cells. The structure of the open channel, vital for understanding the mechanism of channel opening, is incompletely understood. We highlight a new study that unveils the structure of a constitutively active Orai mutant and takes us closer towards understanding the molecular basis of Orai channel activation.


Assuntos
Proteína ORAI1/metabolismo , Animais , Drosophila melanogaster/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ativação do Canal Iônico , Proteína ORAI1/química , Proteína ORAI1/ultraestrutura , Conformação Proteica
9.
J Biol Chem ; 296: 100224, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33361160

RESUMO

The initial activation step in the gating of ubiquitously expressed Orai1 calcium (Ca2+) ion channels represents the activation of the Ca2+-sensor protein STIM1 upon Ca2+ store depletion of the endoplasmic reticulum. Previous studies using constitutively active Orai1 mutants gave rise to, but did not directly test, the hypothesis that STIM1-mediated Orai1 pore opening is accompanied by a global conformational change of all Orai transmembrane domain (TM) helices within the channel complex. We prove that a local conformational change spreads omnidirectionally within the Orai1 complex. Our results demonstrate that these locally induced global, opening-permissive TM motions are indispensable for pore opening and require clearance of a series of Orai1 gating checkpoints. We discovered these gating checkpoints in the middle and cytosolic extended TM domain regions. Our findings are based on a library of double point mutants that contain each one loss-of-function with one gain-of-function point mutation in a series of possible combinations. We demonstrated that an array of loss-of-function mutations are dominant over most gain-of-function mutations within the same as well as of an adjacent Orai subunit. We further identified inter- and intramolecular salt-bridge interactions of Orai subunits as a core element of an opening-permissive Orai channel architecture. Collectively, clearance and synergistic action of all these gating checkpoints are required to allow STIM1 coupling and Orai1 pore opening. Our results unravel novel insights in the preconditions of the unique fingerprint of CRAC channel activation, provide a valuable source for future structural resolutions, and help to understand the molecular basis of disease-causing mutations.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Ativação do Canal Iônico/genética , Proteínas de Neoplasias/química , Proteína ORAI1/química , Molécula 1 de Interação Estromal/química , Substituição de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Regulação da Expressão Gênica , Genes Reporter , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Lipossomos/química , Lipossomos/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Simulação de Dinâmica Molecular , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Técnicas de Patch-Clamp , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo
10.
Int J Mol Sci ; 21(16)2020 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-32764353

RESUMO

Calcium ions regulate a wide array of physiological functions including cell differentiation, proliferation, muscle contraction, neurotransmission, and fertilization. The endoplasmic reticulum (ER) is the major intracellular Ca2+ store and cellular events that induce ER store depletion (e.g., activation of inositol 1,4,5-triphosphate (IP3) receptors) trigger a refilling process known as store-operated calcium entry (SOCE). It requires the intricate interaction between the Ca2+ sensing stromal interaction molecules (STIM) located in the ER membrane and the channel forming Orai proteins in the plasma membrane (PM). The resulting active STIM/Orai complexes form highly selective Ca2+ channels that facilitate a measurable Ca2+ influx into the cytosol followed by successive refilling of the ER by the sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA). STIM and Orai have attracted significant therapeutic interest, as enhanced SOCE has been associated with several cancers, and mutations in STIM and Orai have been linked to immunodeficiency, autoimmune, and muscular diseases. 2-Aminoethyl diphenylborinate (2-APB) is a known modulator and depending on its concentration can inhibit or enhance SOCE. We have synthesized several novel derivatives of 2-APB, introducing halogen and other small substituents systematically on each position of one of the phenyl rings. Using a fluorometric imaging plate reader (FLIPR) Tetra-based calcium imaging assay we have studied how these structural changes of 2-APB affect the SOCE modulation activity at different compound concentrations in MDA-MB-231 breast cancer cells. We have discovered 2-APB derivatives that block SOCE at low concentrations, at which 2-APB usually enhances SOCE.


Assuntos
Compostos de Boro/farmacologia , Neoplasias da Mama/tratamento farmacológico , Proteínas de Neoplasias/genética , Proteína ORAI1/genética , Molécula 1 de Interação Estromal/genética , Moléculas de Interação Estromal/genética , Animais , Compostos de Boro/síntese química , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteína ORAI1/química , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Molécula 1 de Interação Estromal/antagonistas & inibidores , Moléculas de Interação Estromal/antagonistas & inibidores
11.
Int J Mol Sci ; 21(7)2020 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-32252254

RESUMO

Store-operated heteromeric Orai1/Orai3 channels have been discussed in the context of aging, cancer, and immune cell differentiation. In contrast to homomeric Orai1 channels, they exhibit a different pharmacology upon application of reactive oxygen species (ROS) or 2-aminoethoxydiphenyl borate (2-APB) in various cell types. In endogenous cells, subunit composition and arrangement may vary and cannot be defined precisely. In this study, we used patch-clamp electrophysiology to investigate the 2-APB profile of store-operated and store-independent homomeric Orai1 and heteromeric Orai1/Orai3 concatenated channels with defined subunit compositions. As has been shown previous, one or more Orai3 subunit(s) within the channel result(s) in decreased Ca2+ release activated Ca2+ current (ICRAC). Upon application of 50 µM 2-APB, channels with two or more Orai3 subunits exhibit large outward currents and can be activated by 2-APB independent from storedepletion and/or the presence of STIM1. The number and position of Orai3 subunits within the heteromeric store-operated channel change ion conductivity of 2-APB-activated outward current. Compared to homomeric Orai1 channels, one Orai3 subunit within the channel does not alter 2-APB pharmacology. None of the concatenated channel constructs were able to exactly simulate the complex 2-APB pharmacology observed in prostate cancer cells. However, 2-APB profiles of prostate cancer cells are similar to those of concatenated channels with Orai3 subunit(s). Considering the presented and previous results, this indicates that distinct subtypes of heteromeric SOCE channels may be selectively activated or blocked. In the future, targeting distinct heteromeric SOCE channel subtypes may be the key to tailored SOCE-based therapies.


Assuntos
Canais de Cálcio/metabolismo , Ativação do Canal Iônico , Multimerização Proteica , Canais de Cálcio/química , Linhagem Celular Tumoral , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Potenciais da Membrana , Proteína ORAI1/química , Proteína ORAI1/metabolismo , Ligação Proteica , Multimerização Proteica/efeitos dos fármacos
12.
Cell Physiol Biochem ; 54(2): 252-270, 2020 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-32176842

RESUMO

BACKGROUND/AIMS: Store-operated Ca2+ entry (SOCE) through plasma membrane Ca2+ channel Orai1 is essential for many cellular processes. SOCE, activated by ER Ca2+ store-depletion, relies on the gating function of STIM1 Orai1-activating region SOAR of the ER-anchored Ca2+-sensing protein STIM1. Electrophysiologically, SOCE is characterized as Ca2+ release-activated Ca2+ current (ICRAC). A major regulatory mechanism that prevents deleterious Ca2+ overload is the slow Ca2+-dependent inactivation (SCDI) of ICRAC. Several studies have suggested a role of Ca2+/calmodulin (Ca2+/CaM) in triggering SCDI. However, a direct contribution of STIM1 in regulating Ca2+/CaM-mediated SCDI of ICRAC is as yet unclear. METHODS: The Ca2+/CaM binding to STIM1 was tested by pulling down recombinant GFP-tagged human STIM1 C-terminal fragments on CaM sepharose beads. STIM1 was knocked out by CRISPR/Cas9 technique in HEK293 cells stably overexpressing human Orai1. Store-operated Ca2+ influx was measured using Fluorometric Imaging Plate Reader and whole-cell patch clamp in cells transfected with STIM1 CaM binding mutants. The involvement of Ca2+/CaM in SCDI was investigated by including recombinant human CaM in patch pipette in electrophysiology. RESULTS: Here we identified residues Leu374/Val375 (H1) and Leu390/Phe391 (H2) within SOAR that serve as hydrophobic anchor sites for Ca2+/CaM binding. The bifunctional H2 site is critical for both Orai1 activation and Ca2+/CaM binding. Single residue mutations of Phe391 to less hydrophobic residues significantly diminished SOCE and ICRAC, independent of Ca2+/CaM. Hence, the role of H2 residues in Ca2+/CaM-mediated SCDI cannot be precisely evaluated. In contrast, the H1 site controls exclusively Ca2+/CaM binding and subsequently SCDI, but not Orai1 activation. V375A but not V375W substitution eliminated SCDI of ICRAC caused by Ca2+/CaM, proving a direct role of STIM1 in coordinating SCDI. CONCLUSION: Taken together, we propose a mechanistic model, wherein binding of Ca2+/CaM to STIM1 hydrophobic anchor residues, H1 and H2, triggers SCDI by disrupting the functional interaction between STIM1 and Orai1. Our findings reveal how STIM1, Orai1, and Ca2+/CaM are functionally coordinated to control ICRAC.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Calmodulina/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiologia , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/química , Molécula 1 de Interação Estromal/fisiologia , Sistemas CRISPR-Cas , Canais de Cálcio/genética , Sinalização do Cálcio , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Proteínas de Membrana/metabolismo , Modelos Químicos , Modelos Moleculares , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/química , Proteína ORAI1/genética , Ligação Proteica , Domínios Proteicos , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Regulação para Cima
13.
Curr Drug Targets ; 21(1): 55-75, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31556856

RESUMO

BACKGROUND: Calcium (Ca2+) ion is a major intracellular signaling messenger, controlling a diverse array of cellular functions like gene expression, secretion, cell growth, proliferation, and apoptosis. The major mechanism controlling this Ca2+ homeostasis is store-operated Ca2+ release-activated Ca2+ (CRAC) channels. CRAC channels are integral membrane protein majorly constituted via two proteins, the stromal interaction molecule (STIM) and ORAI. Following Ca2+ depletion in the Endoplasmic reticulum (ER) store, STIM1 interacts with ORAI1 and leads to the opening of the CRAC channel gate and consequently allows the influx of Ca2+ ions. A plethora of studies report that aberrant CRAC channel activity due to Loss- or gain-of-function mutations in ORAI1 and STIM1 disturbs this Ca2+ homeostasis and causes several autoimmune disorders. Hence, it clearly indicates that the therapeutic target of CRAC channels provides the space for a new approach to treat autoimmune disorders. OBJECTIVE: This review aims to provide the key structural and mechanical insights of STIM1, ORAI1 and other molecular modulators involved in CRAC channel regulation. RESULTS AND CONCLUSION: Understanding the structure and function of the protein is the foremost step towards improving the effective target specificity by limiting their potential side effects. Herein, the review mainly focusses on the structural underpinnings of the CRAC channel gating mechanism along with its biophysical properties that would provide the solid foundation to aid the development of novel targeted drugs for an autoimmune disorder. Finally, the immune deficiencies caused due to mutations in CRAC channel and currently used pharmacological blockers with their limitation are briefly summarized.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Canais de Cálcio Ativados pela Liberação de Cálcio/química , Canais de Cálcio Ativados pela Liberação de Cálcio/metabolismo , Proteína ORAI1/química , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/química , Molécula 1 de Interação Estromal/metabolismo , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , Canais de Cálcio Ativados pela Liberação de Cálcio/antagonistas & inibidores , Canais de Cálcio Ativados pela Liberação de Cálcio/genética , Sinalização do Cálcio/fisiologia , Humanos , Proteína ORAI1/genética , Molécula 1 de Interação Estromal/genética
14.
J Gen Physiol ; 152(1)2020 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-31816637

RESUMO

Store-operated Orai1 channels regulate a wide range of cellular functions from gene expression to cell proliferation. Previous studies have shown that gating of Orai1 channels is regulated by the outer pore residues V102 and F99, which together function as a hydrophobic gate to block ion conduction in resting channels. Opening of this gate occurs through a conformational change that moves F99 away from the permeation pathway, leading to pore hydration and ion conduction. In addition to this outer hydrophobic gate, several studies have postulated the presence of an inner gate formed by the basic residues R91, K87, and R83 in the inner pore. These positively charged residues were suggested to block ion conduction in closed channels via mechanisms involving either electrostatic repulsion or steric occlusion by a bound anion plug. However, in contrast to this model, here we find that neutralization of the basic residues dose-dependently abolishes both STIM1-mediated and STIM1-independent activation of Orai1 channels. Molecular dynamics simulations show that loss of the basic residues dehydrates the pore around the hydrophobic gate and stabilizes the pore in a closed configuration. Likewise, the severe combined immunodeficiency mutation, Orai1 R91W, closes the channel by dewetting the hydrophobic stretch of the pore and stabilizing F99 in a pore-facing configuration. Loss of STIM1-gating in R91W and in the other basic residue mutants is rescued by a V102A mutation, which restores pore hydration at the hydrophobic gate to repermit ion conduction. These results indicate that the inner pore basic residues facilitate opening of the principal outer hydrophobic gate through a long-range effect involving hydration of the outer pore.


Assuntos
Substituição de Aminoácidos , Ativação do Canal Iônico , Proteína ORAI1/química , Arginina/química , Arginina/genética , Células HEK293 , Humanos , Lisina/química , Lisina/genética , Simulação de Dinâmica Molecular , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Domínios Proteicos
15.
Hum Mol Genet ; 29(11): 1808-1823, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-31600783

RESUMO

Loss-of function mutations in Orai1 Ca2+ channels lead to a form of severe combined immunodeficiency, auto-immunity, muscle hypotonia and defects in dental enamel production and sweat gland function. Two single-nucleotide polymorphisms (SNPs) in Orai1 have been found and localize to the second extracellular loop. These polymorphisms associate with atopic dermatitis but how they affect Ca2+ signalling and cell function is unknown. Here, we find that Orai1-SNPs turnover considerably more slowly than wild type Orai1 and are more abundantly expressed in the plasma membrane. We show a central role for flotillin in the endocytotic recycling of Orai1 channels and that endocytosed wild type Orai1 is trafficked to Rab 7-positive late endosomes for lysosomal degradation. Orai1-SNPs escape the degradation pathway and instead enter Rab 11-positive recycling endosomes, where they are returned to the surface membrane through Arf6-dependent exocytosis. We find that Orai1-SNPs escape late endosomes through endosomal pH regulation of interaction between the channel and flotillin. We identify a pH-sensitive electrostatic interaction between positively charged arginine in extracellular loop 2 (K210) and a negatively charged aspartate (D112) in extracellular loop 1 that helps determine Orai1 turnover. The increase in membrane Orai1-SNP leads to a mis-match in Orai1-STIM stoichiometry, resulting in inhibition of Ca2+ entry and Ca2+-dependent gene expression. Our results identify new strategies for targeting atopic dermatitis.


Assuntos
Cálcio/metabolismo , Dermatite Atópica/genética , Proteína ORAI1/genética , Proteínas rab de Ligação ao GTP/genética , Cálcio/química , Sinalização do Cálcio/genética , Membrana Celular/química , Membrana Celular/genética , Dermatite Atópica/patologia , Endossomos/genética , Regulação da Expressão Gênica/genética , Células HEK293 , Humanos , Lisossomos/genética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteína ORAI1/química , Polimorfismo de Nucleotídeo Único/genética , Proteólise , proteínas de unión al GTP Rab7
16.
Sci Signal ; 12(608)2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31744929

RESUMO

The stromal interaction molecule 1 (STIM1) has two important functions, Ca2+ sensing within the endoplasmic reticulum and activation of the store-operated Ca2+ channel Orai1, enabling plasma-membrane Ca2+ influx. We combined molecular dynamics (MD) simulations with live-cell recordings and determined the sequential Ca2+-dependent conformations of the luminal STIM1 domain upon activation. Furthermore, we identified the residues within the canonical and noncanonical EF-hand domains that can bind to multiple Ca2+ ions. In MD simulations, a single Ca2+ ion was sufficient to stabilize the luminal STIM1 complex. Ca2+ store depletion destabilized the two EF hands, triggering disassembly of the hydrophobic cleft that they form together with the stable SAM domain. Point mutations associated with tubular aggregate myopathy or cancer that targeted the canonical EF hand, and the hydrophobic cleft yielded constitutively clustered STIM1, which was associated with activation of Ca2+ entry through Orai1 channels. On the basis of our results, we present a model of STIM1 Ca2+ binding and refine the currently known initial steps of STIM1 activation on a molecular level.


Assuntos
Cálcio/metabolismo , Simulação de Dinâmica Molecular , Proteínas de Neoplasias/química , Domínios Proteicos , Desdobramento de Proteína , Molécula 1 de Interação Estromal/química , Algoritmos , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Motivos EF Hand , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Microscopia Confocal , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/química , Proteína ORAI1/metabolismo , Ratos , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo
17.
Biochem Biophys Res Commun ; 516(4): 1066-1072, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31279526

RESUMO

Intracellular Ca2+ signals play many important cellular functions such as migration, proliferation and differentiation. Store-operated Ca2+ entry (SOCE) is a major route of Ca2+ entry in nonexcitable cells. The activation of SOCE requires engagement between stromal interaction molecule 1 (STIM1) molecules on the endoplasmic reticulum and Ca2+ release-activated Ca2+ (CRAC) channel Orais (Orai1-3) on the plasma membrane. Accumulating evidence indicates that SOCE plays critical roles in cancer cell proliferation, invasion and metastasis. Here, we used the synthetic intracellular peptides derived from the C-termini of Orai channels to treat the breast cancer cells. We have found that Orai3-CT peptide exhibits stronger binding to STIM1 than Orai1-CT, and Orai3-CT peptide acts in a dominant negative fashion, blocking the STIM1-Orai1 interaction and reducing the Ca2+ entry and proliferation of breast cancer cells.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Canais de Cálcio Ativados pela Liberação de Cálcio/farmacologia , Proliferação de Células/efeitos dos fármacos , Peptídeos/farmacologia , Antineoplásicos/química , Neoplasias da Mama/metabolismo , Cálcio/metabolismo , Canais de Cálcio/química , Canais de Cálcio/farmacologia , Canais de Cálcio Ativados pela Liberação de Cálcio/química , Sinalização do Cálcio/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/química , Proteína ORAI1/farmacologia , Peptídeos/química , Mapas de Interação de Proteínas/efeitos dos fármacos , Molécula 1 de Interação Estromal/metabolismo
18.
SAR QSAR Environ Res ; 30(2): 81-108, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30773908

RESUMO

Upregulation of store-operated Ca2+ influx via ORAI1, an integral component of the CRAC channel, is responsible for abnormal cytokine release in active rheumatoid arthritis, and therefore ORAI1 has been proposed as an attractive molecular target. In this study, we attempted to predict the mechanical insights of ORAI1 inhibitors through pharmacophore modelling, 3D-QSAR, molecular docking and free energy analysis. Various hypotheses of pharmacophores were generated and from that, a pharmacophore hypothesis with two hydrogen bond acceptors, one hydrogen bond donor and two aromatic rings (AADRR) resulted in a statistically significant 3D-QSAR model (r2 = 0.84 and q2 = 0.74). We believe that the obtained statistical model is a reliable QSAR model for the diverse dataset of inhibitors against the IL-2 production assay. The visualization of contours in active and inactive compounds generated from the 3D-QSAR models and molecular docking studies revealed major interaction with GLN108, HIS113 and ASP114, and interestingly, these residues are located near the Ca2+ selectivity filter region. Free energy binding analysis revealed that Coulomb energy, van der Waals energy and non-polar solvation terms are more favourable for ligand binding. Thus, the present study provides the physical and chemical requirements for the development of novel ORAI1 inhibitors with improved biological activity.


Assuntos
Simulação de Acoplamento Molecular , Proteína ORAI1/química , Relação Quantitativa Estrutura-Atividade , Ligação de Hidrogênio
19.
Cell Calcium ; 79: 35-43, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30807904

RESUMO

Substantial progress has been made in the past several years in establishing the stoichiometries of STIM and Orai proteins and understanding their influence on store-operated calcium entry. Depletion of ER Ca2+ triggers STIM1 to accumulate at ER-plasma membrane junctions where it binds and opens Ca2+ release-activated Ca2+ (CRAC) channels. STIM1 is a dimer, and release of Ca2+ from its two luminal domains is reported to promote their association as well as drive formation of higher-order STIM1 oligomers. The CRAC channel, originally thought to be tetrameric, is now considered to be a hexamer of Orai1 subunits based on crystallographic and electrophysiological studies. STIM1 binding activates CRAC channels in a highly nonlinear way, such that all six Orai1 binding sites must be occupied to account for the activation and signature properties of native channels. The structural basis of STIM1 engagement with the channel is currently unclear, with evidence suggesting that STIM1 dimers bind to individual or pairs of Orai1 subunits. This review examines evidence that has led to points of consensus and debate about STIM1 and Orai1 stoichiometries, and explains the importance of STIM-Orai complex stoichiometry for the regulation of store-operated calcium entry.


Assuntos
Cálcio/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Cálcio/química , Canais de Cálcio Ativados pela Liberação de Cálcio/metabolismo , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Proteínas de Neoplasias/química , Proteína ORAI1/química , Molécula 1 de Interação Estromal/química
20.
Curr Opin Cell Biol ; 57: 90-98, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30716649

RESUMO

Store-operated calcium entry (SOCE) through Orai channels is triggered by receptor-stimulated depletion of Ca2+ from the ER. Orai1 is unique in terms of its activation mechanism, biophysical properties, and structure, and its precise regulation is essential for human health. Recent studies have begun to reveal the structural basis of the major steps in the SOCE pathway and how the system is reliably suppressed in resting cells but able to respond robustly to ER Ca2+ depletion. In this review, we discuss current models describing the activation of ER Ca2+ sensor STIM1, its binding to Orai1, propagation of the binding signal from the channel periphery to the central pore, and the resulting conformational changes underlying opening of the highly Ca2+ selective Orai1 channel.


Assuntos
Cálcio/metabolismo , Proteína ORAI1/química , Molécula 1 de Interação Estromal/química , Animais , Sinalização do Cálcio , Humanos , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...