Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
DNA Repair (Amst) ; 133: 103593, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38029688

RESUMEN

To maintain tissue homeostasis, cell proliferation is balanced by cell death. PARP1 is an important protein involved in both processes. Upon sensing DNA damage, PARP1 forms poly(ADP-ribose) (PAR) chains to recruit the repair proteins, ensuring genome integrity and faithful cell proliferation. In addition, PAR also regulates the activity of PARP1. Persistent DNA damage can signal the cell to progress toward programmed cell death, apoptosis. During apoptosis, proteolytic cleavage of PARP1 generates an N-terminal, ZnF1-2PARP1 (DNA binding or regulatory fragment), and C-terminal, PARP1ΔZnF1-2 (catalytic or PAR carrier fragment), which exhibits a basal activity. Regulation of the apoptotic fragments by PAR has not been studied. Here, we report that PAR inhibits the basal level activity of PARP1ΔZnF1-2, and ZnF1-2PARP1 interacts with PARP1ΔZnF1-2 to exhibit DNA-dependent stimulation and partially restores the PAR-dependent stimulation. Interestingly, along with the auto-modification domain of PARP1, the DNA-binding domains, ZnF1-2PARP1, also acts as an acceptor of PARylation; therefore, ZnF1-2PARP1 exhibits a reduced affinity for DNA upon PARylation. Furthermore, we show that ZnF1-2PARP1 shows trans-dominant inhibition of DNA-dependent stimulation of PARP2. Altogether, our study explores the regulation of the catalytic activity of PARP1ΔZnF1-2 and PARP2 by the regulatory apoptotic fragment of PARP1.


Asunto(s)
ADN , Poli Adenosina Difosfato Ribosa , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , ADN/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Poli ADP Ribosilación , Reparación del ADN , Daño del ADN
2.
Comput Struct Biotechnol J ; 21: 5003-5015, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37867971

RESUMEN

Modular proteins are regulatory proteins that carry out more than one function. These proteins upregulate or downregulate a biochemical cascade to establish homeostasis in cells. To switch the function or alter the efficiency (based on cellular needs), these proteins require different facilitators that bind to a site different from the catalytic (active/orthosteric) site, aka 'allosteric site', and fine-tune their function. These facilitators (or effectors) are allosteric modulators. In this Review, we have discussed the allostery, characterized them based on their mechanisms, and discussed how allostery plays an important role in the activity modulation and function fine-tuning of proteins. Recently there is an emergence in the discovery of allosteric drugs. We have also emphasized the role, significance, and future of allostery in therapeutic applications.

3.
FEBS J ; 290(21): 5098-5113, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37462479

RESUMEN

Poly(ADP-ribosyl)ation is predominantly catalyzed by Poly(ADP-ribose) polymerase 1 (PARP1) in response to DNA damage, mediating the DNA repair process to maintain genomic integrity. Single-strand (SSB) and double-strand (DSB) DNA breaks are bona fide stimulators of PARP1 activity. However, PAR-mediated PARP1 regulation remains unexplored. Here, we report ZnF3, BRCT, and WGR, hitherto uncharacterized, as PAR reader domains of PARP1. Surprisingly, these domains recognize PARylated protein with a higher affinity compared with PAR but bind with weak or no affinity to DNA breaks as standalone domains. Conversely, ZnF1 and ZnF2 of PARP1 recognize DNA breaks but bind weakly to PAR. In addition, PAR reader domains, together, exhibit a synergy to recognize PAR or PARylated protein. Further competition-binding studies suggest that PAR binding releases DNA from PARP1, and the WGR domain facilitates DNA release. Unexpectedly, PAR showed catalytic stimulation of PARP1 but hampered the DNA-dependent stimulation. Altogether, our work discovers dedicated high-affinity PAR reader domains of PARP1 and uncovers a novel mechanism of allosteric regulation of DNA-dependent and DNA-independent activities of PARP1 by its catalytic product PAR.


Asunto(s)
Reparación del ADN , Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/genética , ADN/metabolismo , Daño del ADN , Poli Adenosina Difosfato Ribosa/metabolismo
4.
FEBS J ; 290(18): 4533-4542, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37246313

RESUMEN

PARP1 is a nuclear protein involved in the maintenance of genomic stability. It catalyses the formation of poly(ADP-ribose) (PAR) to recruit repair proteins at the site of DNA lesions, such as double-strand and single-strand breaks. In the process of DNA replication or repair, there could occur stretch of ssDNA, usually protected by ssDNA binding proteins, but when present in abundance can turn into DNA beaks and cause cell death. PARP1 is an extremely sensitive sensor of DNA breaks; however, the interaction of PARP1 with single-stranded DNA (ssDNA) remains unexplored. Here, we report that the two Zn-fingers, ZnF1 and ZnF2, of PARP1, mediate high-affinity recognition of ssDNA. Our studies suggest that although PAR and ssDNA are chemical analogues, they are recognized by a distinct set of domains of PARP1, yet PAR not only induces dislodging of ssDNA from PARP1 but also hampers the ssDNA-dependent PARP1 activity. It is noteworthy that PAR carrier apoptotic fragment PARP1ΔZnF1-2 gets cleaved from PARP1 to facilitate apoptosis, leaving behind the DNA-bound ZnF1-ZnF2PARP1 . Our studies demonstrate that the PARP1ΔZnF1-2 is competent for ssDNA-dependent stimulation only in the presence of another apoptotic fragment ZnF1-ZnF2PARP1 , suggesting the indispensability of DNA-bound ZnF1-ZnF2PARP1 dual domains for the same.


Asunto(s)
ADN de Cadena Simple , Poli(ADP-Ribosa) Polimerasas , Animales , Poli(ADP-Ribosa) Polimerasas/metabolismo , ADN de Cadena Simple/genética , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli Adenosina Difosfato Ribosa/metabolismo , ADN/metabolismo , Reparación del ADN
5.
FEBS J ; 290(4): 1060-1077, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36128736

RESUMEN

Su-(var)3-9 homologue 5 (SUVH5), a member of SUVH family of histone lysine methyltransferase (HKMT) in Arabidopsis, is involved in epigenetic regulation of chromatin by recognizing 5-methyl-cytosine (5mC), in both CpG and non-CpG DNA context, through SRA domain and simultaneously performing the di-methylation of lysine 9 of histone H3 (H3K9) through SET domain. Here, we establish that the SET domain of SUVH5 allosterically restricts the SRA domain to the 5mC containing strand(s) of fully methylated CpG, hemi-methylated CpG and methylated CpHpH DNA. In addition, SET domain enhances the binding affinity of the SRA-SET dual domains to fully-mCpG but not to hemi-mCpG. Also, the recognition of methylated DNA by the SRA positively influences the recognition of H3K9 by the SET domain. Our further studies revealed that the SET domain recognizes the "A(R/K)KST" motif present in H3K9 and in other histone H2A variants. Further, computational analyses and quantum mechanics/molecular mechanics calculations explain the bases for robust mono-MTase but weak di-MTase activities of SUVH5. Given that the majority of eukaryotic proteins, including those involved in epigenetic gene regulation, contain more than one domain, our study suggests that understanding the allosteric regulation among multiple domains of proteins is relevant for unravelling biological outcomes.


Asunto(s)
Arabidopsis , Histonas , Metiltransferasas , Regulación Alostérica , Arabidopsis/metabolismo , ADN/metabolismo , Metilación de ADN , Epigénesis Genética , Histonas/genética , Histonas/metabolismo , Lisina/metabolismo , Metiltransferasas/metabolismo , Dominios PR-SET
6.
J Phys Chem B ; 126(45): 9137-9151, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36326054

RESUMEN

An increased level of naturally occurring anti-TDP-43 antibodies was observed in the serum and cerebrospinal fluid (CSF) of amyotrophic lateral sclerosis patients. Human serum albumin (HSA), the most abundant protein in blood plasma and CSF, is found to interact with pathological proteins like Aß and α-synuclein. Therefore, we examined the effect on the in vitro aggregation of a C-terminal fragment of TDP-43 in the presence of HSA. We found that the lag phase in TDP-432C aggregation is abrogated in the presence of HSA, but there is an overall decreased aggregation as examined by thioflavin-T fluorescence spectroscopy and microscopy. An early onset of TDP-432C oligomer formation in the presence of HSA was observed using atomic force microscopy and transmission electron microscopy. Also, a known chemical inhibitor of TDP-432Caggregation, AIM4, abolishes the HSA-induced early formation of TDP-432C oligomers. Notably, the aggregates of TDP-432C formed in the presence of HSA are more stable against sarkosyl detergent. Using affinity copurification, we observed that HSA can bind to TDP-432C, and biolayer interferometry further supported their physical interaction and suggested the binding affinity to be in sub-micromolar range. Taken together, the data support that HSA can interact with TDP-432C in vitro and affect its aggregation.


Asunto(s)
Esclerosis Amiotrófica Lateral , Albúmina Sérica Humana , Humanos , Esclerosis Amiotrófica Lateral/metabolismo , Microscopía de Fuerza Atómica , Agregación Patológica de Proteínas
7.
Int J Pharm ; 613: 121358, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-34896560

RESUMEN

Ideal dressing materials for complex and large asymmetric burns should have the dual properties of anti-bacterial and regenerative with advanced applicability of direct deposit on the wound at the patient bedside. In this study, core-shell nanofibers (polycaprolactone; PCL and polyethylene oxide; PEO) with different percent of silver sulfadiazine (SSD) loading (2-10%) were prepared by the airbrushing method using a custom build device. Results indicate a sustained release profile of silver sulfadiazine (SSD) up to 28 days and concentration-dependent anti-bacterial activity. The morphology and proliferation of human dermal fibroblast (HDF) cells and human dental follicle stem cells (HDFSC) on the silver sulfadiazine loaded nanofibers confirm the biocompatibility of airbrushed nanofibers. Moreover, upregulation of extracellular matrix (ECM) proteins (Col I, Col III, and elastin) support the differentiation and regenerative properties of silver sulfadiazine nanofiber mats. This was further confirmed by the complete recovery of rabbit burn wound models within 7 days of silver sulfadiazine loaded nanofiber dressing. Histopathology data show silver sulfadiazine loaded core-shell nanofibers' anti-inflammatory and proliferative activity without any adverse response on the tissue. Overall data display that the airbrushed silver sulfadiazine-loaded core-shell nanofibers are effective dressing material with the possibility of direct fiber deposition on the wound to cover, heal, and regenerate large asymmetric burn wounds.


Asunto(s)
Quemaduras , Nanofibras , Animales , Vendajes , Quemaduras/tratamiento farmacológico , Humanos , Conejos , Sulfadiazina de Plata , Cicatrización de Heridas
8.
Biochemistry ; 60(35): 2652-2662, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34404204

RESUMEN

The PHD finger-containing VARIANT IN METHYLATION/ORTHRUS (VIM/ORTH) family of proteins in Arabidopsis consists of functional homologues of mammalian UHRF1 and is required for the maintenance of DNA methylation. Comparison of the sequence with those of other PHD fingers implied that VIM1 and VIM3 PHD could recognize lysine 4 of histone H3 (H3K4) through interactions mediated by a conserved aspartic acid. However, our calorimetric and modified histone peptide array binding studies suggested that neither H3K4 nor other histone marks are recognized by VIM1 and VIM3 PHD fingers. Here, we report a 2.6 Å resolution crystal structure of the VIM1 PHD finger and demonstrate significant structural changes in the putative H3 recognition segments in contrast to canonical H3K4 binding PHD fingers. These changes include (i) the H3A1 binding region, (ii) strand ß1 that forms an intermolecular ß-sheet with the H3 peptide, and (iii) an aspartate-containing motif involved in salt bridge interaction with H3K4, which together appear to abrogate recognition of H3K4 by the VIM1 PHD finger. To understand the significance of the altered structural features in the VIM1 PHD that might prevent histone H3 recognition, we modeled a chimeric VIM1 PHD (chmVIM1 PHD) by grafting the peptide binding structural features of the BHC80 PHD onto the VIM1 PHD. Molecular dynamics simulation and metadynamics analyses revealed that the chmVIM1 PHD-H3 complex is stable and also showed a network of intermolecular interactions similar to those of the BHC80 PHD-H3 complex. Collectively, this study reveals that subtle structural changes in the peptide binding region of the VIM1 PHD abrogate histone H3 recognition.


Asunto(s)
Proteínas de Arabidopsis/química , Arabidopsis/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/química , Histonas/metabolismo , Dedos de Zinc PHD , Fragmentos de Péptidos/química , Secuencia de Aminoácidos , Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Sitios de Unión , Proteínas de Unión al ADN/metabolismo , Conformación Molecular , Simulación de Dinámica Molecular , Fragmentos de Péptidos/metabolismo , Homología de Secuencia
9.
Sci Rep ; 11(1): 12293, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112881

RESUMEN

Phosphodiesterase (PDE) inhibitors, such as pentoxifylline (PTX), are used as pharmacological agents to enhance sperm motility in assisted reproductive technology (ART), mainly to aid the selection of viable sperm in asthenozoospermic ejaculates and testicular spermatozoa, prior to intracytoplasmic sperm injection (ICSI). However, PTX is reported to induce premature acrosome reaction (AR) and, exert toxic effects on oocyte function and early embryo development. Additionally, in vitro binding studies as well as computational binding free energy (ΔGbind) suggest that PTX exhibits weak binding to sperm PDEs, indicating room for improvement. Aiming to reduce the adverse effects and to enhance the sperm motility, we designed and studied PTX analogues. Using structure-guided in silico approach and by considering the physico-chemical properties of the binding pocket of the PDEs, designed analogues of PTX. In silico assessments indicated that PTX analogues bind more tightly to PDEs and form stable complexes. Particularly, ex vivo evaluation of sperm treated with one of the PTX analogues (PTXm-1), showed comparable beneficial effect at much lower concentration-slower AR, higher DNA integrity and extended longevity of  spermatozoa and  superior embryo quality. PTXm-1 is proposed to be a better pharmacological agent for ART than PTX for sperm function enhancement.


Asunto(s)
Astenozoospermia/tratamiento farmacológico , Pentoxifilina/química , Hidrolasas Diéster Fosfóricas/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Acrosoma/efectos de los fármacos , Astenozoospermia/patología , Humanos , Masculino , Estructura Molecular , Pentoxifilina/análogos & derivados , Pentoxifilina/farmacología , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/química , Técnicas Reproductivas Asistidas/tendencias , Inyecciones de Esperma Intracitoplasmáticas/métodos , Espermatozoides/crecimiento & desarrollo , Testículo/efectos de los fármacos , Testículo/patología
10.
Int J Biol Macromol ; 170: 514-522, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33359809

RESUMEN

Non-CpG DNA methylation (non-mCpG) is enriched in the genome of brain neurons and germline cells in mammals. Accumulation of non-mCpG during postnatal brain development correlates with gene regulation and inactivation of distal regulatory elements. Recently, UHRF1 has been found to contribute to de novo non-CpG methylation, however, whether UHRF1 could recognize non-mCpG is unknown. Here, we have demonstrated through calorimetric measurements that the UHRF1 SRA can recognize mCpH and fully-mCpHpG, types of non-mCpG. Our ITC binding studies endorse the preferential reading of hemi-mCpG by UHRF1 SRA and also show 6-fold weaker binding for fully-mCpG than hemi-mCpG. Despite presence of symmetrical (5-methyl cytosine) 5mCs, stoichiometry of 1:1 for UHRF1 SRA binding to fully-mCpG indicates that UHRF1 SRA may not form a stable complex with fully-mCpG DNA. Contrarily, UHRF1 SRA recognizes fully-mCpHpG with a stoichiometry of 2:1 protein to DNA duplex with binding affinity higher than fully-mCpG. Our crystal structure of UHRF1 SRA bound to fully-mCpHpG DNA reveals dual flip-out mechanism of 5mC recognition. Metadynamics studies corroborates with ITC data that UHRF1 SRA could not form a stable complex with fully-mCpG DNA. Altogether, this study demonstrates that UHRF1 SRA recognizes non-mCpG DNA and exhibits contrasting mechanisms for hemi-mCpG and fully-mCpHpG DNA recognition.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Islas de CpG/fisiología , Citosina/metabolismo , Metilación de ADN/fisiología , ADN/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , 5-Metilcitosina/metabolismo , Secuencia de Aminoácidos , Animales , ADN Complementario/metabolismo , Fosfatos de Dinucleósidos/metabolismo , Ratones , Unión Proteica/fisiología , Alineación de Secuencia
11.
Int J Biol Macromol ; 147: 117-130, 2020 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-31917988

RESUMEN

TDP-43 is an RNA/DNA-binding protein which is also implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS) disease. TDP-43's cytoplasmic mis-localization, liquid-liquid phase separation (LLPS) due to RNA depletion and aggregation, are proposedly important TDP-43-toxicity causing mechanisms. So far, therapeutic options for ALS are extremely ineffective hence, multi-faceted approaches such as targeting the oxidative stress and inhibiting the TDP-43's aggregation, are being actively pursued. Recently, we have identified an acridine derivative, AIM4, as an anti-TDP-43 aggregation molecule however, its mechanism is not deciphered. Here, we have utilized computational tools to examine binding site(s) of AIM4 in the TDP-43 structure and compared with other relevant compounds. We find that AIM4 has a binding site in the C-terminal amyloidogenic region (aa: 288-319), with Gly-288 & Phe-289 residues which are also important for TDP-43's LLPS. Importantly, alike to previously reported effects of RNA, AIM4 could also inhibit the in vitro LLPS of a C-terminal fragment TDP-432C bearing an A315T familial mutation. Furthermore, isothermal titration calorimetry (ITC) data also support the binding of AIM4 to TDP-432C-A315T. This antagonism of AIM4 towards TDP-43's LLPS and presence of binding site of AIM4 on TDP-43 support AIM4's potential to be an important molecule towards ALS therapeutic research.


Asunto(s)
Acridinas/química , Esclerosis Amiotrófica Lateral/metabolismo , Simulación por Computador , Proteínas de Unión al ADN/química , Agregado de Proteínas , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Proteínas Mutantes/química , Conformación Proteica , Estabilidad Proteica , Termodinámica
12.
ACS Omega ; 4(5): 9593-9602, 2019 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-31460050

RESUMEN

In all the living systems, reactive oxygen species (ROS) metabolism provides resistance against internal and external oxidative stresses. Auranofin (AF), an FDA-approved gold [Au(I)]-conjugated drug, is known to selectively target thiol-reductases, key enzymes involved in ROS metabolism. AF has been successfully tested for its inhibitory activity through biochemical studies, both in vitro and in vivo, against a diverse range of pathogens including protozoa, nematodes, bacteria, and so forth. Cocrystal structures of thiol-reductases complexed with AF revealed that Au(I) was coordinately linked to catalytic cysteines, but the mechanism of transfer of Au(I) from AF to catalytic cysteines still remains unknown. In this study, we have employed computational approaches to understand the interaction of AF with thiol-reductases of selected human pathogens. A similar network of interactions of AF was observed in all the studied enzymes. Also, we have shown that tailor-made analogues of AF can be designed against selective thiol-reductases for targeted inhibition. Molecular dynamics studies show that the AF-intermediates, tetraacetylthioglucose (TAG)-gold, and triethylphosphine (TP)-gold, coordinately linked to one of catalytic cysteines, remain stable in the binding pocket of thiol-reductases for Leishmania infantum and Plasmodium falciparum (PfTrxR). This suggests that the TP and TAG moieties of AF may be sequentially eliminated during the transfer of Au(I) to catalytic cysteines of the receptor.

13.
Biochimie ; 149: 105-114, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29656054

RESUMEN

UHRF1 is a multi-domain protein comprising of a tandem tudor (UHRF1 TTD), a PHD finger, and a SET and RING-associated domain. It is required for the maintenance of CG methylation, heterochromatin formation and DNA repair. Isothermal titration calorimetry binding studies of unmodified and methylated lysine histone peptides establish that the UHRF1 TTD binds dimethylated Lys9 on histone H3 (H3K9me2). Further, MD simulation and binding studies reveal that TTD-PHD of UHRF1 (UHRF1 TTD-PHD) preferentially recognizes dimethyl-lysine status. Importantly, we show that Asp145 in the binding pocket determines the preferential recognition of the dimethyl-ammonium group of H3K9me2. Interestingly, PHD finger of the UHRF1 TTD-PHD has a negligible contribution to the binding affinity for recognition of K9me2 by the UHRF1 TTD. Surprisingly, Lys4 methylation on H3 peptide has an insignificant effect on combinatorial recognition of R2 and K9me2 on H3 by the UHRF1 TTD-PHD. We propose that subtle variations of key residues at the binding pocket determine status specific recognition of histone methyl-lysines by the reader domains.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/química , Metilación de ADN/genética , N-Metiltransferasa de Histona-Lisina/química , Dominios Proteicos , Sitios de Unión/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Calorimetría , Reparación del ADN/genética , Escherichia coli/genética , Heterocromatina/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/química , Histonas/genética , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Proteína 1 de Unión al Supresor Tumoral P53/química , Proteína 1 de Unión al Supresor Tumoral P53/genética , Ubiquitina-Proteína Ligasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...