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










Base de datos
Intervalo de año de publicación
1.
Structure ; 32(5): 594-602.e4, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38460521

RESUMEN

Apoptosis-inducing factor (AIF), which is confined to mitochondria of normal healthy cells, is the first identified caspase-independent cell death effector. Moreover, AIF is required for the optimal functioning of the respiratory chain machinery. Recent findings have revealed that AIF fulfills its pro-survival function by interacting with CHCHD4, a soluble mitochondrial protein which promotes the entrance and the oxidative folding of different proteins in the inner membrane space. Here, we report the crystal structure of the ternary complex involving the N-terminal 27-mer peptide of CHCHD4, NAD+, and AIF harboring its FAD (flavin adenine dinucleotide) prosthetic group in oxidized form. Combining this information with biophysical and biochemical data on the CHCHD4/AIF complex, we provide a detailed structural description of the interaction between the two proteins, validated by both chemical cross-linking mass spectrometry analysis and site-directed mutagenesis.


Asunto(s)
Factor Inductor de la Apoptosis , Dominio Catalítico , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales , Modelos Moleculares , Unión Proteica , Factor Inductor de la Apoptosis/metabolismo , Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/genética , Humanos , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Regulación Alostérica , Cristalografía por Rayos X , NAD/metabolismo , NAD/química , Sitios de Unión , Factores de Transcripción/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética
2.
J Zhejiang Univ Sci B ; 24(2): 172-184, 2023 Feb 15.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-36751702

RESUMEN

Auditory neuropathy spectrum disorder (ANSD) represents a variety of sensorineural deafness conditions characterized by abnormal inner hair cells and/or auditory nerve function, but with the preservation of outer hair cell function. ANSD represents up to 15% of individuals with hearing impairments. Through mutation screening, bioinformatic analysis and expression studies, we have previously identified several apoptosis-inducing factor (AIF) mitochondria-associated 1 (AIFM1) variants in ANSD families and in some other sporadic cases. Here, to elucidate the pathogenic mechanisms underlying each AIFM1 variant, we generated AIF-null cells using the clustered regularly interspersed short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system and constructed AIF-wild type (WT) and AIF-mutant (mut) (p.|T260A, p.|R422W, and p.|R451Q) stable transfection cell lines. We then analyzed AIF structure, coenzyme-binding affinity, apoptosis, and other aspects. Results revealed that these variants resulted in impaired dimerization, compromising AIF function. The reduction reaction of AIF variants had proceeded slower than that of AIF-WT. The average levels of AIF dimerization in AIF variant cells were only 34.5%|‒|49.7% of that of AIF-WT cells, resulting in caspase-independent apoptosis. The average percentage of apoptotic cells in the variants was 12.3%|‒|17.9%, which was significantly higher than that (6.9%|‒|7.4%) in controls. However, nicotinamide adenine dinucleotide (NADH) treatment promoted the reduction of apoptosis by rescuing AIF dimerization in AIF variant cells. Our findings show that the impairment of AIF dimerization by AIFM1 variants causes apoptosis contributing to ANSD, and introduce NADH as a potential drug for ANSD treatment. Our results help elucidate the mechanisms of ANSD and may lead to the provision of novel therapies.


Asunto(s)
Factor Inductor de la Apoptosis , NAD , Humanos , Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , NAD/metabolismo , Dimerización , Apoptosis
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34548399

RESUMEN

Combining mass spectrometry-based chemical cross-linking and complexome profiling, we analyzed the interactome of heart mitochondria. We focused on complexes of oxidative phosphorylation and found that dimeric apoptosis-inducing factor 1 (AIFM1) forms a defined complex with ∼10% of monomeric cytochrome c oxidase (COX) but hardly interacts with respiratory chain supercomplexes. Multiple AIFM1 intercross-links engaging six different COX subunits provided structural restraints to build a detailed atomic model of the COX-AIFM12 complex (PDBDEV_00000092). An application of two complementary proteomic approaches thus provided unexpected insight into the macromolecular organization of the mitochondrial complexome. Our structural model excludes direct electron transfer between AIFM1 and COX. Notably, however, the binding site of cytochrome c remains accessible, allowing formation of a ternary complex. The discovery of the previously overlooked COX-AIFM12 complex and clues provided by the structural model hint at potential roles of AIFM1 in oxidative phosphorylation biogenesis and in programmed cell death.


Asunto(s)
Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/metabolismo , Apoptosis , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Mitocondrias Cardíacas/metabolismo , Membranas Mitocondriales/metabolismo , Fosforilación Oxidativa , Animales , Bovinos , Transporte de Electrón , Conformación Proteica
4.
J Med Chem ; 64(15): 11445-11459, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34338510

RESUMEN

Blocking the interaction between the apoptosis-inducing factor (AIF) and cyclophilin A (CypA) by the AIF fragment AIF(370-394) is protective against glutamate-induced neuronal cell death and brain injury in mice. Starting from AIF(370-394), we report the generation of the disulfide-bridged and shorter variant AIF(381-389) and its structural characterization by nuclear magnetic resonance (NMR) in the free and CypA-bound state. AIF(381-389) in both the free and bound states assumes a ß-hairpin conformation similar to that of the fragment in the AIF protein and shows a highly reduced conformational flexibility. This peptide displays a similar in vitro affinity for CypA, an improved antiapoptotic activity in cells and an enhanced proteolytic stability compared to the parent peptide. The NMR-based 3D model of the AIF(381-389)/CypA complex provides a better understanding of the binding hot spots on both the peptide and the protein and can be exploited to design AIF/CypA inhibitors with improved pharmacokinetic and pharmacodynamics features.


Asunto(s)
Factor Inductor de la Apoptosis/farmacología , Apoptosis/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Ciclofilina A/antagonistas & inhibidores , Diseño de Fármacos , Animales , Factor Inductor de la Apoptosis/síntesis química , Factor Inductor de la Apoptosis/química , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ciclofilina A/metabolismo , Relación Dosis-Respuesta a Droga , Ácido Glutámico/metabolismo , Humanos , Ratones , Estructura Molecular , Relación Estructura-Actividad
5.
Methods Mol Biol ; 2280: 189-198, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33751436

RESUMEN

To perform their action usually flavoproteins interact transiently with a variety of molecular partners, whose binding is reciprocally affected and often controlled by the redox state of the bound flavin cofactor. As a case study, here we describe an approach for the quantitative characterization of the redox-controlled interaction of the mammalian apoptosis inducing factor (AIF) with one of its known protein partners, namely, the mitochondrial coiled-coil-helix-coiled-coil-helix domain-containing protein 4 (CHCHD4). In particular, we report a protocol for the titration of the flavoprotein in both in its oxidized and reduced states with CHCHD4, using an implementation of the MicroScale Thermophoresis (MST) technique.


Asunto(s)
Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/metabolismo , Escherichia coli/crecimiento & desarrollo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Regulación Alostérica , Anaerobiosis , Animales , Factor Inductor de la Apoptosis/genética , Escherichia coli/genética , Fluorescencia , Ratones , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Oxidación-Reducción , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espectrofotometría
6.
Oxid Med Cell Longev ; 2021: 6673661, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33510840

RESUMEN

The human apoptosis-inducing factor (hAIF) is a moonlight flavoprotein involved in mitochondrial respiratory complex assembly and caspase-independent programmed cell death. These functions might be modulated by its redox-linked structural transition that enables hAIF to act as a NAD(H/+) redox sensor. Upon reduction with NADH, hAIF undergoes a conformational reorganization in two specific insertions-the flexible regulatory C-loop and the 190-202 ß-harpin-promoting protein dimerization and the stabilization of a long-life charge transfer complex (CTC) that modulates its monomer-dimer equilibrium and its protein interaction network in healthy mitochondria. In this regard, here, we investigated the precise function of the ß-hairpin in the AIF conformation landscape related to its redox mechanism, by analyzing the role played by W196, a key residue in the interaction of this motif with the regulatory C-loop. Mutations at W196 decrease the compactness and stability of the oxidized hAIF, indicating that the ß-hairpin and C-loop coupling contribute to protein stability. Kinetic studies complemented with computational simulations reveal that W196 and the ß-hairpin conformation modulate the low efficiency of hAIF as NADH oxidoreductase, contributing to configure its active site in a noncompetent geometry for hydride transfer and to stabilize the CTC state by enhancing the affinity for NAD+. Finally, the ß-hairpin motif contributes to define the conformation of AIF's interaction surfaces with its physiological partners. These findings improve our understanding on the molecular basis of hAIF's cellular activities, a crucial aspect for clarifying its associated pathological mechanisms and developing new molecular therapies.


Asunto(s)
Factor Inductor de la Apoptosis/química , NAD/química , Secuencias de Aminoácidos , Factor Inductor de la Apoptosis/genética , Humanos , Oxidación-Reducción , Conformación Proteica en Lámina beta , Estabilidad Proteica
7.
IUBMB Life ; 73(3): 568-581, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33035389

RESUMEN

In Homo sapiens, the apoptosis-inducing factor (AIF) family is represented by three different proteins, known as AIF, AMID and AIFL, that have in common the mitochondrial localisation in healthy cells, the presence of FAD- and NADH-dependent domains involved in an -albeit yet not well understood- oxidoreductase function and their capability to induce programmed cell death. AIF is the best characterised family member, while the information about AMID and AIFL is much scarcer. Nonetheless, available data support different roles as well as mechanisms of action of their particular apoptogenic and redox domains regarding both pro-apoptotic and anti-apoptotic activities. Moreover, diverse cellular functions, to date far from fully clarified, are envisaged for the transcripts corresponding to these three proteins. Here, we review the so far available knowledge on the moonlighting human AIF family from their molecular properties to their relevance in health and disease, through the evaluation of their potential cell death and redox functions in their different subcellular locations. This picture emerging from the current knowledge of the AIF family envisages its contribution to regulate signalling and transcription machineries in the crosstalk among mitochondria, the cytoplasm and the nucleus.


Asunto(s)
Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/metabolismo , Núcleo Celular/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Animales , Apoptosis , Factor Inductor de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Muerte Celular , Humanos , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Familia de Multigenes , Neoplasias/metabolismo , Neoplasias/patología , Filogenia
8.
Arch Biochem Biophys ; 692: 108515, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32791141

RESUMEN

Apoptosis-inducing factor (AIF) is a flavoprotein and essential partner of the CHCHD4 redox protein during the mitochondrial intermembrane space import machinery. Mammalian AIF has three cysteine residues, which have received little attention. Previous reports have evidenced a redox interaction between AIF and thioredoxin 1 (Trx1), particularly after oxidant conditions. Therefore, we asked whether the cysteine residues of the human AIF could be oxidized. Our data showed that endogenous AIF could be oxidized to disulfide-linked conjugates (DLC). Overexpressed WT AIF in HEK293T cells, as well as recombinant WT AIF, formed DLC. Expression of C256S, C317S or C441S AIF mutants severely inhibited DLC formation in cells exposed to oxidants. In vitro, DLC formation was completely precluded with C256S and C441S AIF mutants and partially inhibited with the C317S mutant. DLC was shown to enhance cellular susceptibility to apoptosis induced by staurosporine, likely by preventing AIF to maintain mitochondrial oxidative phosphorylation. Cells with decreased expression of Trx1 produced more AIF DLC than those with normal Trx1 levels, and in vitro, Trx1 was able to decrease the amount of AIF DLC. Finally, confocal analysis, as well as immunoblotting of mitochondrial fraction, indicated that a fraction of Trx1 is present in mitochondria. Overall, these data provide evidence that all three cysteine residues of AIF can be oxidized to DLC, which can be disrupted by mitochondrial Trx1.


Asunto(s)
Factor Inductor de la Apoptosis , Apoptosis , Disulfuros , Sustitución de Aminoácidos , Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Células HEK293 , Células HeLa , Humanos , Mutación Missense , Oxidación-Reducción , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estaurosporina/farmacología
9.
Biochim Biophys Acta Gen Subj ; 1864(12): 129717, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32861757

RESUMEN

BACKGROUND: The neuronal apoptotic process requires the nuclear translocation of Apoptosis Inducing Factor (AIF) in complex with Cyclophilin A (CypA) with consequent chromatin condensation and DNA degradation events. Targeting CypA by delivering an AIF-blocking peptide (AIF(370-394)) provides a significant neuroprotection, demonstrating the biological relevance of the AIF/CypA complex. To date pharmaceutical compounds targeting this complex are missing. METHODS: We designed and synthesized a set of mono and bicyclic AIF(370-394) analogs containing both disulfide and 1,2,3-triazole bridges, in the attempt to both stabilize the peptide conformation and improve its binding affinity to CypA. Peptide structures in solution and in complex with CypA have been studied by circular dichroism (CD), Nuclear Magnetic Resonance (NMR) and molecular modeling. The ability of stapled peptides to interact with CypA was evaluated by using Epic Corning label free technique and Isothermal Titration Calorimetry experiments. RESULTS: We identified a stapled peptide analogue of AIF(370-394) with a ten-fold improved affinity for CypA. Molecular modeling studies reveal that the new peptide acquires ß-turn/ß-fold structures and shares with the parent molecule the same binding region on CypA. CONCLUSIONS: Data obtained provide invaluable assistance in designing new ligand of CypA for therapeutic approaches in neurodegenerative diseases. GENERAL SIGNIFICANCE: Due to the crucial role of AIF/CypA complex formation in neurodegeneration, identification of selective inhibitors is of high importance for targeted therapies. We describe new bicyclic peptide inhibitors with improved affinity for CypA, investigating the kinetic, thermodynamic and structural effects of conformational constraints on the protein-ligand interaction, and their utility for drug design.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Ciclofilina A/metabolismo , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Péptidos/química , Péptidos/farmacología , Factor Inductor de la Apoptosis/química , Diseño de Fármacos , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Fármacos Neuroprotectores/síntesis química , Péptidos/síntesis química
10.
BMC Cancer ; 19(1): 451, 2019 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-31088422

RESUMEN

BACKGROUND: In a time of increasing concerns over personalized and precision treatment in breast cancer (BC), filtering prognostic factors attracts more attention. Apoptosis-Inducing Factor Mitochondrion-associated 3 (AIFM3) is widely expressed in various tissues and aberrantly expressed in several cancers. However, clinical implication of AIFM3 has not been reported in BC. The aim of the study is to investigate the crystal structure, clinical and prognostic implications of AIFM3 in BC. METHODS: AIFM3 expression in 151 BC samples were assessed by immunohistochemistry (IHC). The Cancer Genome Atlas (TCGA) and Kaplan-Meier survival analysis were used to demonstrate expression and survival of AIFM3 signature. Gene Set Enrichment Analysis (GSEA) was performed to investigate the mechanisms related to AIFM3 expression in BC. RESULTS: AIFM3 was significantly more expressed in breast cancer tissues than in normal tissues. AIFM3 expression had a significant association with tumor size, lymph node metastasis, TNM stage and molecular typing. Higher AIFM3 expression was related to a shorter overall survival (OS) and disease-free survival (DFS). Lymph node metastasis and TNM stage were independent factors of AIFM3 expression. The study presented the crystal structure of AIFM3 successfully and predicted several binding sites when AIFM3 bonded to PTPN12 by Molecular Operating Environment software (MOE). CONCLUSIONS: AIFM3 might be a potential biomarker for predicting prognosis in BC, adding to growing evidence that AIFM3 might interact with PTPN12.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , Neoplasias de la Mama/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Regulación hacia Arriba , Adulto , Anciano , Anciano de 80 o más Años , Factor Inductor de la Apoptosis/química , Sitios de Unión , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática , Persona de Mediana Edad , Proteínas Mitocondriales/química , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estadificación de Neoplasias , Pronóstico , Carga Tumoral
11.
Antioxid Redox Signal ; 30(18): 2013-2029, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-30450916

RESUMEN

Aims: The human apoptosis-inducing factor (hAIF) supports OXPHOS biogenesis and programmed cell death, with missense mutations producing neurodegenerative phenotypes. hAIF senses the redox environment of cellular compartments, stabilizing a charge transfer complex (CTC) dimer that modulates the protein interaction network. In this context, we aimed to evaluate the subcellular pH, CTC formation, and pathogenic mutations effects on hAIF stability, and a thermal denaturation high-throughput screening (HTS) assay to discover AIF binders. Results: Apoptotic hAIFΔ1-101 is not stable at intermembrane mitochondrial space (IMS) pH, but the 77-101 residues confer stability to the mitochondrial isoform. hAIF and its CTC populate different conformational ensembles with redox switch to the CTC producing a less stable and compact protein. The pathogenic G308E, ΔR201, and E493V mutations modulate hAIF stability; particularly, ΔR201 causes a population shift to a less stable conformation that remodels active site structure and dynamics. We have identified new molecules that modulate the hAIF reduced nicotinamide adenine dinucleotide (NADH)/oxidized nicotinamide adenine dinucleotide (NAD+) association/dissociation equilibrium and regulate its catalytic efficiency. Innovation: Biophysical methods allow evaluating the regulation of hAIF functional ensembles and to develop an HTS assay to discover small molecules that might modulate hAIF stability and activities. Conclusions: The mitochondrial soluble 54-77 portion stabilizes hAIF at the IMS pH. NADH-redox-linked conformation changes course with strong NAD+ binding and protein dimerization, but they produce a negative impact in overall hAIF stability. Loss of functionality in the R201 deletion is due to distortion of the active site architecture. We report molecules that may serve as leads in the development of hAIF bioactive compounds.


Asunto(s)
Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/metabolismo , Mutación , Factor Inductor de la Apoptosis/genética , Muerte Celular , Humanos , Concentración de Iones de Hidrógeno , Mitocondrias/metabolismo , Modelos Moleculares , NAD/metabolismo , Oxidación-Reducción , Unión Proteica , Conformación Proteica , Multimerización de Proteína
12.
Biochem J ; 475(14): 2377-2393, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-29891613

RESUMEN

The complex formation between the proteins apoptosis-inducing factor (AIF) and cyclophilin A (CypA) following oxidative stress in neuronal cells has been suggested as a main target for reverting ischemia-stroke damage. Recently, a peptide encompassing AIF residues 370-394 has been developed to target the AIF-binding site on CypA, to prevent the association between the two proteins and suppress glutamate-induced cell death in neuronal cells. Using a combined approach based on NMR spectroscopy, synthesis and in vitro testing of all Ala-scan mutants of the peptide and molecular docking/molecular dynamics, we have generated a detailed model of the AIF (370-394)/CypA complex. The model suggests us that the central region of the peptide spanning residues V374-K384 mostly interacts with the protein and that for efficient complex inhibition and preservation of CypA activity, it is bent around amino acids F46-G75 of the protein. The model is consistent with experimental data also from previous works and supports the concept that the peptide does not interfere with other CypA activities unrelated to AIF activation; therefore, it may serve as an ideal template for generating future non-peptidic antagonists.


Asunto(s)
Factor Inductor de la Apoptosis/química , Ciclofilina A/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Péptidos/química , Humanos , Espectroscopía de Resonancia Magnética
13.
Acta Histochem ; 120(4): 385-394, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29685720

RESUMEN

Programmed cell death is a fundamental event that takes place during organ development and plays an important role in cellular homeostasis. Since various body organs of the camel are under high ecological and physiological stress during food and water deprivation, desiccation, and the long exposure to solar radiation in these desert nomads, we aimed to examine the immunohistochemical expression of apoptosis-related biomarkers in some of its normal body organs to illustrate a basic track for further pathological investigation. Regarding apoptosis, the present study has revealed that the higher expression of cleaved caspase-9 (CC9) [initiator of the intrinsic pathway] and CC3 (effector caspase), and the scanty expression of CC8 (initiator of the extrinsic pathway), highlight the role of the caspase-dependent, intrinsic apoptotic pathway particularly in the intestines and lymphoid organs. The apoptosis- inducing factor (AIF)-immunoexpression was completely missing in the cell nuclei of the examined tissues, indicating the absence of the caspase-independent pathway. The nuclear overexpression of the phospho-histone H2AX (γ H2AX) and the occasional expression of single-stranded DNA, particularly among the CNS neurons, suggest an efficient, protective DNA-repair mechanism in such cells. Thus, despite efficient anti-apoptotic mechanisms intrinsic apoptotic pathways exists in brain, intestine and lymph organs of adult desert camels.


Asunto(s)
Factor Inductor de la Apoptosis/química , Apoptosis , Biomarcadores/química , Animales , Factor Inductor de la Apoptosis/metabolismo , Camelus , Caspasa 3/química , Inmunohistoquímica , Modelos Animales
14.
Methods Enzymol ; 592: 417-455, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28668129

RESUMEN

DNA damage outcomes depend upon the efficiency and fidelity of DNA damage responses (DDRs) for different cells and damage. As such, DDRs represent tightly regulated prototypical systems for linking nanoscale biomolecular structure and assembly to the biology of genomic regulation and cell signaling. However, the dynamic and multifunctional nature of DDR assemblies can render elusive the correlation between the structures of DDR factors and specific biological disruptions to the DDR when these structures are altered. In this chapter, we discuss concepts and strategies for combining structural, biophysical, and imaging techniques to investigate DDR recognition and regulation, and thus bridge sequence-level structural biochemistry to quantitative biological outcomes visualized in cells. We focus on representative DDR responses from PARP/PARG/AIF damage signaling in DNA single-strand break repair and nonhomologous end joining complexes in double-strand break repair. Methods with exemplary experimental results are considered with a focus on strategies for probing flexibility, conformational changes, and assembly processes that shape a predictive understanding of DDR mechanisms in a cellular context. Integration of structural and imaging measurements promises to provide foundational knowledge to rationally control and optimize DNA damage outcomes for synthetic lethality and for immune activation with resulting insights for biology and cancer interventions.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Daño del ADN , Reparación del ADN , Glicósido Hidrolasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Transducción de Señal , Animales , Factor Inductor de la Apoptosis/química , Microscopía por Crioelectrón/métodos , Cristalografía por Rayos X/métodos , ADN/química , ADN/genética , ADN/metabolismo , Inestabilidad Genómica , Glicósido Hidrolasas/química , Humanos , Microscopía Fluorescente/métodos , Modelos Moleculares , Neoplasias/genética , Neoplasias/metabolismo , Imagen Óptica/métodos , Poli(ADP-Ribosa) Polimerasas/química
15.
Biochem Biophys Res Commun ; 490(3): 1011-1017, 2017 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-28666871

RESUMEN

The apoptosis-inducing factor (AIF) is a FAD-containing protein playing critical roles in caspase-independent apoptosis and mitochondrial respiratory chain biogenesis and maintenance. While its lethal role is well known, the details of its mitochondrial function remain elusive. So far, nineteen allelic variants of AIF have been associated to human diseases, mainly affecting the nervous system. A strict correlation is emerging between the degree of impairment of its ability to stabilize the charge-transfer (CT) complex between FAD and NAD+ and the severity of the resulting pathology. Recently, we demonstrated that the G307E replacement in murine AIF (equivalent to the pathogenic G308E in the human protein) dramatically decreases the rate of CT complex formation through the destabilization of the flavoprotein interaction with NAD(H). To provide further insights into the structural bases of its altered functional properties, here we report the first crystal structure of an AIF pathogenic mutant variant in complex with NAD+ (murine AIF-G307ECT) in comparison with its oxidized form. With respect to wild type AIF, the mutation leads to an altered positioning of NAD+ adenylate moiety, which slows down CT complex formation. Moreover, the altered balance between the binding of the adenine/nicotinamide portions of the coenzyme determines a large drop in AIF-G307E ability to discriminate between NADH and NADPH.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , NADP/metabolismo , NAD/metabolismo , Mutación Puntual , Animales , Factor Inductor de la Apoptosis/química , Cristalografía por Rayos X , Ratones , Modelos Moleculares , Conformación Proteica , Especificidad por Sustrato
16.
Sci Rep ; 7(1): 1138, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28442737

RESUMEN

The Cyclophilin A (CypA)/Apoptosis Inducing Factor (AIF) complex is implicated in the DNA degradation in response to various cellular stress conditions, such as oxidative stress, cerebral hypoxia-ischemia and traumatic brain injury. The pro-apoptotic form of AIF (AIF(Δ1-121)) mainly interacts with CypA through the amino acid region 370-394. The AIF(370-394) synthetic peptide inhibits complex formation in vitro by binding to CypA and exerts neuroprotection in a model of glutamate-mediated oxidative stress. Here, the binding site of AIF(Δ1-121) and AIF(370-394) on CypA has been mapped by NMR spectroscopy and biochemical studies, and a molecular model of the complex has been proposed. We show that AIF(370-394) interacts with CypA on the same surface recognized by AIF(Δ1-121) protein and that the region is very close to the CypA catalytic pocket. Such region partially overlaps with the binding site of cyclosporin A (CsA), the strongest catalytic inhibitor of CypA. Our data point toward distinct CypA structural determinants governing the inhibitor selectivity and the differential biological effects of AIF and CsA, and provide new structural insights for designing CypA/AIF selective inhibitors with therapeutic relevance in neurodegenerative diseases.


Asunto(s)
Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/metabolismo , Ciclofilina A/química , Ciclofilina A/metabolismo , Sitios de Unión , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Unión Proteica , Mapeo de Interacción de Proteínas
17.
Structure ; 24(12): 2067-2079, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27818101

RESUMEN

Apoptosis-inducing factor (AIF) is critical for mitochondrial respiratory complex biogenesis and for mediating necroptotic parthanatos; these functions are seemingly regulated by enigmatic allosteric switching driven by NADH charge-transfer complex (CTC) formation. Here, we define molecular pathways linking AIF's active site to allosteric switching regions by characterizing dimer-permissive mutants using small-angle X-ray scattering (SAXS) and crystallography and by probing AIF-CTC communication networks using molecular dynamics simulations. Collective results identify two pathways propagating allostery from the CTC active site: (1) active-site H454 links to S480 of AIF's central ß-strand to modulate a hydrophobic border at the dimerization interface, and (2) an interaction network links AIF's FAD cofactor, central ß-strand, and Cß-clasp whereby R529 reorientation initiates C-loop release during CTC formation. This knowledge of AIF allostery and its flavoswitch mechanism provides a foundation for biologically understanding and biomedically controlling its participation in mitochondrial homeostasis and cell death.


Asunto(s)
Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/metabolismo , NAD/metabolismo , Regulación Alostérica , Dominio Catalítico , Cristalografía por Rayos X , Dimerización , Humanos , Modelos Moleculares , Estructura Secundaria de Proteína , Dispersión del Ángulo Pequeño , Difracción de Rayos X
18.
Fish Shellfish Immunol ; 56: 84-95, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27368536

RESUMEN

Apoptosis inducing factor (AIF) and cytochrome c (CYC) are two mitochondrial apoptogenic factors. In the present study, the cDNA sequences of AIF (LvAIF) and CYC (LvCYC) were cloned from Pacific white shrimp, Litopenaeus vannamei. The LvAIF was 1664 bp, including a 5'-terminal untranslated region (UTR) of 154 bp, an open reading frame (ORF) of 1323 bp encoding a polypeptide of 440 amino acids (aa) and a 3' UTR of 187 bp. The LvCYC was 582 bp, including a 50 bp 5' UTR, a 315 bp ORF encoding for 104 aa, and a 217 bp 3' UTR. The deduced protein of LvAIF contained a conserved Pyr_redox and AIF_C domain at the N-terminal and the predicted LvCYC included a conservative cytochrome_C domain, respectively. Phylogenetic analysis revealed that LvAIF belonged to AIF1 subfamily and showed a close relationship with AIF1 from vertebrates and LvCYC showed the closest relationship with its counterparts from shrimp Marsupenaeus japonicus. Tissue expression profiles showed that both LvAIF and LvCYC existed in most tissues, with the most predominant expression of LvAIF in intestine, then followed muscle and the weakest expression in gill. The highest expression of LvCYC was detected in muscle, and the weakest expression was in hemocytes. Additionally, after white spot syndrome virus (WSSV) infection, the significant up-regulation of LvAIF, LvCYC and caspase 3 transcripts and the increase of pro-caspase 3 and active-caspase 3 protein were detected at most time points (P < 0.05). However, all of the three genes down-regulated in hemocytes in the early stage after WSSV infection. WSSV proliferation and shrimp mortality showed a time-dependent manner and the production of ROS in hemocytes were significantly increased at 6 and 24 h after infection. Our results showed that the apoptotic genes AIF, CYC and caspase 3 might play crucial roles in hepatopancreas, however, the production of ROS in hemocytes might be important in shrimp defense against WSSV infection.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Proteínas de Artrópodos/genética , Citocromos c/genética , Regulación de la Expresión Génica , Inmunidad Innata , Penaeidae/genética , Penaeidae/inmunología , Secuencia de Aminoácidos , Animales , Factor Inductor de la Apoptosis/química , Factor Inductor de la Apoptosis/metabolismo , Proteínas de Artrópodos/química , Proteínas de Artrópodos/metabolismo , Secuencia de Bases , Clonación Molecular , Citocromos c/química , Citocromos c/metabolismo , ADN Complementario/genética , ADN Complementario/metabolismo , Hemocitos/inmunología , Hemocitos/virología , Hepatopáncreas/inmunología , Hepatopáncreas/virología , Penaeidae/metabolismo , Penaeidae/virología , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Alineación de Secuencia , Virus del Síndrome de la Mancha Blanca 1/fisiología
19.
J Mol Biol ; 428(18): 3650-65, 2016 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-27178839

RESUMEN

The X-linked AIFM1 gene encodes mitochondrial apoptosis-inducing factor (AIF), an FAD-containing and NADH-specific oxidoreductase critically important for energy metabolism and execution of the caspase-independent cell death pathway. Several recently identified mutations in human AIFM1 lead to neurodegenerative disorders varying in severity and onset time. This study was undertaken to structurally and functionally characterize four pathologic variants of human AIF: V243L, G262S, G308E, and G338E. A strong correlation between the mutational effects on the AIF function and clinical phenotype was observed only for the G308E aberration, drastically damaging both the redox properties of AIF and mitochondrial respiration. In contrast, only minimal or mild changes were detected in the structure/function of AIF V243L and G338E, respectively, indicating that a marked decrease in their cellular expression likely triggers the disease. Alterations in the structure and redox activity of AIF G262S, on the other hand, were more severe than could be predicted based on the clinical phenotype. Together, the results of this and previous studies allow to conclude that the phenotypic variability and severity of the AIFM1-related disorders depend on which AIF feature is predominantly affected (i.e., cellular production level, structure, redox or apoptogenic function) and to what extent. Only a drastic decrease in the expression level or/and redox activity of AIF tends to cause an early and severe neurodegeneration, whereas less pronounced changes in the AIF properties could lead to a broad range of slowly progressive neurological disorders.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Factor Inductor de la Apoptosis/química , Expresión Génica , Humanos , Cinética , Modelos Moleculares , Proteínas Mutantes/química , Oxidación-Reducción , Conformación Proteica
20.
Eur J Hum Genet ; 24(3): 463-6, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26173962

RESUMEN

AIFM1 is a gene located on the X chromosome, coding for AIF (Apoptosis-Inducing Factor), a mitochondrial flavoprotein involved in caspase-independent cell death. AIFM1 mutations have been associated with different clinical phenotypes: a severe infantile encephalopathy with combined oxidative phosphorylation deficiency and the Cowchock syndrome, an X-linked Charcot-Marie-Tooth disease (CMTX4) with axonal sensorimotor neuropathy, deafness and cognitive impairment. In two male cousins with early-onset mitochondrial encephalopathy and cytochrome c oxidase (COX) deficiency, we identified a novel AIFM1 mutation. Muscle biopsies and electromyography in both patients showed signs of severe denervation. Our patients manifested a phenotype that included signs of both cortical and motor neuron involvement. These observations emphasize the role of AIF in the development and function of neurons.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Predisposición Genética a la Enfermedad , Enfermedad de la Neurona Motora/genética , Mutación/genética , Secuencia de Aminoácidos , Factor Inductor de la Apoptosis/química , Familia , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Fenotipo , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...