Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.457
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 184(17): 4531-4546.e26, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34314702

RESUMEN

Defects in translation lead to changes in the expression of proteins that can serve as drivers of cancer formation. Here, we show that cytosolic NAD+ synthesis plays an essential role in ovarian cancer by regulating translation and maintaining protein homeostasis. Expression of NMNAT-2, a cytosolic NAD+ synthase, is highly upregulated in ovarian cancers. NMNAT-2 supports the catalytic activity of the mono(ADP-ribosyl) transferase (MART) PARP-16, which mono(ADP-ribosyl)ates (MARylates) ribosomal proteins. Depletion of NMNAT-2 or PARP-16 leads to inhibition of MARylation, increased polysome association and enhanced translation of specific mRNAs, aggregation of their translated protein products, and reduced growth of ovarian cancer cells. Furthermore, MARylation of the ribosomal proteins, such as RPL24 and RPS6, inhibits polysome assembly by stabilizing eIF6 binding to ribosomes. Collectively, our results demonstrate that ribosome MARylation promotes protein homeostasis in cancers by fine-tuning the levels of protein synthesis and preventing toxic protein aggregation.


Asunto(s)
ADP-Ribosilación , Neoplasias Ováricas/metabolismo , Biosíntesis de Proteínas , Proteostasis , Ribosomas/metabolismo , Regiones no Traducidas 3'/genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Estrés del Retículo Endoplásmico , Trompas Uterinas/metabolismo , Femenino , Humanos , Ratones Endogámicos NOD , Ratones SCID , NAD/metabolismo , Nicotinamida-Nucleótido Adenililtransferasa , Conformación de Ácido Nucleico , Neoplasias Ováricas/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Polirribosomas/metabolismo , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Ribosómicas/metabolismo
2.
Annu Rev Biochem ; 85: 431-54, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26844395

RESUMEN

Recent developments indicate that macrodomains, an ancient and diverse protein domain family, are key players in the recognition, interpretation, and turnover of ADP-ribose (ADPr) signaling. Crucial to this is the ability of macrodomains to recognize ADPr either directly, in the form of a metabolic derivative, or as a modification covalently bound to proteins. Thus, macrodomains regulate a wide variety of cellular and organismal processes, including DNA damage repair, signal transduction, and immune response. Their importance is further indicated by the fact that dysregulation or mutation of a macrodomain is associated with several diseases, including cancer, developmental defects, and neurodegeneration. In this review, we summarize the current insights into macrodomain evolution and how this evolution influenced their structural and functional diversification. We highlight some aspects of macrodomain roles in pathobiology as well as their emerging potential as therapeutic targets.


Asunto(s)
Reparación del ADN , Proteínas de Escherichia coli/química , Neoplasias/enzimología , Poli(ADP-Ribosa) Polimerasas/química , Procesamiento Proteico-Postraduccional , Proteínas Represoras/química , Virosis/enzimología , Adenosina Difosfato Ribosa/química , Adenosina Difosfato Ribosa/metabolismo , Animales , Daño del ADN , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Evolución Molecular , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Familia de Multigenes , Neoplasias/química , Neoplasias/genética , Neoplasias/patología , Filogenia , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Dominios Proteicos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Homología Estructural de Proteína , Virosis/genética , Virosis/patología , Virosis/virología
3.
Mol Cell ; 83(10): 1552-1572, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-37119811

RESUMEN

PARPs catalyze ADP-ribosylation-a post-translational modification that plays crucial roles in biological processes, including DNA repair, transcription, immune regulation, and condensate formation. ADP-ribosylation can be added to a wide range of amino acids with varying lengths and chemical structures, making it a complex and diverse modification. Despite this complexity, significant progress has been made in developing chemical biology methods to analyze ADP-ribosylated molecules and their binding proteins on a proteome-wide scale. Additionally, high-throughput assays have been developed to measure the activity of enzymes that add or remove ADP-ribosylation, leading to the development of inhibitors and new avenues for therapy. Real-time monitoring of ADP-ribosylation dynamics can be achieved using genetically encoded reporters, and next-generation detection reagents have improved the precision of immunoassays for specific forms of ADP-ribosylation. Further development and refinement of these tools will continue to advance our understanding of the functions and mechanisms of ADP-ribosylation in health and disease.


Asunto(s)
ADP-Ribosilación , Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/metabolismo , Procesamiento Proteico-Postraduccional , Adenosina Difosfato Ribosa/metabolismo
4.
Mol Cell ; 82(16): 2939-2951.e5, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35793673

RESUMEN

PARP1 rapidly detects DNA strand break damage and allosterically signals break detection to the PARP1 catalytic domain to activate poly(ADP-ribose) production from NAD+. PARP1 activation is characterized by dynamic changes in the structure of a regulatory helical domain (HD); yet, there are limited insights into the specific contributions that the HD makes to PARP1 allostery. Here, we have determined crystal structures of PARP1 in isolated active states that display specific HD conformations. These captured snapshots and biochemical analysis illustrate HD contributions to PARP1 multi-domain and high-affinity interaction with DNA damage, provide novel insights into the mechanics of PARP1 allostery, and indicate how HD active conformations correspond to alterations in the catalytic region that reveal the active site to NAD+. Our work deepens the understanding of PARP1 catalytic activation, the dynamics of the binding site of PARP inhibitor compounds, and the mechanisms regulating PARP1 retention on DNA damage.


Asunto(s)
Daño del ADN , NAD , Dominio Catalítico , Reparación del ADN , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Poli Adenosina Difosfato Ribosa , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología
5.
Mol Cell ; 82(5): 969-985.e11, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35182479

RESUMEN

Poly(ADP-ribose) (PAR) is an RNA-like polymer that regulates an increasing number of biological processes. Dysregulation of PAR is implicated in neurodegenerative diseases characterized by abnormal protein aggregation, including amyotrophic lateral sclerosis (ALS). PAR forms condensates with FUS, an RNA-binding protein linked with ALS, through an unknown mechanism. Here, we demonstrate that a strikingly low concentration of PAR (1 nM) is sufficient to trigger condensation of FUS near its physiological concentration (1 µM), which is three orders of magnitude lower than the concentration at which RNA induces condensation (1 µM). Unlike RNA, which associates with FUS stably, PAR interacts with FUS transiently, triggering FUS to oligomerize into condensates. Moreover, inhibition of a major PAR-synthesizing enzyme, PARP5a, diminishes FUS condensation in cells. Despite their structural similarity, PAR and RNA co-condense with FUS, driven by disparate modes of interaction with FUS. Thus, we uncover a mechanism by which PAR potently seeds FUS condensation.


Asunto(s)
Esclerosis Amiotrófica Lateral , Poli Adenosina Difosfato Ribosa , Esclerosis Amiotrófica Lateral/genética , Humanos , Poli Adenosina Difosfato Ribosa/metabolismo , ARN/genética , Proteína FUS de Unión a ARN/metabolismo
6.
Mol Cell ; 82(7): 1359-1371.e9, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35216668

RESUMEN

The chromatin-binding protein 53BP1 promotes DNA repair by orchestrating the recruitment of downstream effectors including PTIP, RIF1, and shieldin to DNA double-strand break sites. While we know how PTIP recognizes 53BP1, the molecular details of RIF1 recruitment to DNA-damage sites remains undefined. Here, we report that RIF1 is a phosphopeptide-binding protein that directly interacts with three phosphorylated 53BP1 epitopes. The RIF1-binding sites on 53BP1 share an essential LxL motif followed by two closely apposed phosphorylated residues. Simultaneous mutation of these sites on 53BP1 abrogates RIF1 accumulation into ionizing-radiation-induced foci, but surprisingly, only fully compromises 53BP1-dependent DNA repair when an alternative mode of shieldin recruitment to DNA-damage sites is also disabled. Intriguingly, this alternative mode of recruitment still depends on RIF1 but does not require its interaction with 53BP1. RIF1 therefore employs phosphopeptide recognition to promote DNA repair but also modifies shieldin action independently of 53BP1 binding.


Asunto(s)
Fosfopéptidos , Proteínas de Unión a Telómeros , Proteína BRCA1/genética , Proteínas Portadoras/metabolismo , ADN/metabolismo , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Fosfopéptidos/genética , Fosfopéptidos/metabolismo , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
7.
Mol Cell ; 82(11): 2032-2049.e7, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35460603

RESUMEN

Virus infection modulates both host immunity and host genomic stability. Poly(ADP-ribose) polymerase 1 (PARP1) is a key nuclear sensor of DNA damage, which maintains genomic integrity, and the successful application of PARP1 inhibitors for clinical anti-cancer therapy has lasted for decades. However, precisely how PARP1 gains access to cytoplasm and regulates antiviral immunity remains unknown. Here, we report that DNA virus induces a reactive nitrogen species (RNS)-dependent DNA damage and activates DNA-dependent protein kinase (DNA-PK). Activated DNA-PK phosphorylates PARP1 on Thr594, thus facilitating the cytoplasmic translocation of PARP1 to inhibit the antiviral immunity both in vitro and in vivo. Mechanistically, cytoplasmic PARP1 interacts with and directly PARylates cyclic GMP-AMP synthase (cGAS) on Asp191 to inhibit its DNA-binding ability. Together, our findings uncover an essential role of PARP1 in linking virus-induced genome instability with inhibition of host immunity, which is of relevance to cancer, autoinflammation, and other diseases.


Asunto(s)
Antivirales , Nucleotidiltransferasas , Antivirales/farmacología , Citoplasma/genética , Citoplasma/metabolismo , ADN , Daño del ADN , Inestabilidad Genómica , Humanos , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo
8.
Mol Cell ; 82(12): 2315-2334, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35271815

RESUMEN

ADP-ribosylation (ADPRylation) is a post-translational modification of proteins catalyzed by ADP-ribosyl transferase (ART) enzymes, including nuclear PARPs (e.g., PARP1 and PARP2). Historically, studies of ADPRylation and PARPs have focused on DNA damage responses in cancers, but more recent studies elucidate diverse roles in a broader array of biological processes. Here, we summarize the expanding array of molecular mechanisms underlying the biological functions of nuclear PARPs with a focus on PARP1, the founding member of the family. This includes roles in DNA repair, chromatin regulation, gene expression, ribosome biogenesis, and RNA biology. We also present new concepts in PARP1-dependent regulation, including PAR-dependent post-translational modifications, "ADPR spray," and PAR-mediated biomolecular condensate formation. Moreover, we review advances in the therapeutic mechanisms of PARP inhibitors (PARPi) as well as the progress on the mechanisms of PARPi resistance. Collectively, the recent progress in the field has yielded new insights into the expanding universe of PARP1-mediated molecular and therapeutic mechanisms in a variety of biological processes.


Asunto(s)
ADP-Ribosilación , Reparación del ADN , Cromatina/genética , Daño del ADN , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Procesamiento Proteico-Postraduccional , ARN/metabolismo
9.
Mol Cell ; 81(7): 1515-1533.e5, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33571423

RESUMEN

Loss of the ataxia-telangiectasia mutated (ATM) kinase causes cerebellum-specific neurodegeneration in humans. We previously demonstrated that deficiency in ATM activation via oxidative stress generates insoluble protein aggregates in human cells, reminiscent of protein dysfunction in common neurodegenerative disorders. Here, we show that this process is driven by poly-ADP-ribose polymerases (PARPs) and that the insoluble protein species arise from intrinsically disordered proteins associating with PAR-associated genomic sites in ATM-deficient cells. The lesions implicated in this process are single-strand DNA breaks dependent on reactive oxygen species, transcription, and R-loops. Human cells expressing Mre11 A-T-like disorder mutants also show PARP-dependent aggregation identical to ATM deficiency. Lastly, analysis of A-T patient cerebellum samples shows widespread protein aggregation as well as loss of proteins known to be critical in human spinocerebellar ataxias that is not observed in neocortex tissues. These results provide a hypothesis accounting for loss of protein integrity and cerebellum function in A-T.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/deficiencia , Roturas del ADN de Cadena Simple , Proteína Homóloga de MRE11/deficiencia , Neocórtex/metabolismo , Poli ADP Ribosilación , Proteostasis , Ataxias Espinocerebelosas/metabolismo , Adulto , Línea Celular Tumoral , Femenino , Humanos , Masculino , Neocórtex/patología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología
10.
Genes Dev ; 34(5-6): 263-284, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32029451

RESUMEN

ADP-ribosylation is an intricate and versatile posttranslational modification involved in the regulation of a vast variety of cellular processes in all kingdoms of life. Its complexity derives from the varied range of different chemical linkages, including to several amino acid side chains as well as nucleic acids termini and bases, it can adopt. In this review, we provide an overview of the different families of (ADP-ribosyl)hydrolases. We discuss their molecular functions, physiological roles, and influence on human health and disease. Together, the accumulated data support the increasingly compelling view that (ADP-ribosyl)hydrolases are a vital element within ADP-ribosyl signaling pathways and they hold the potential for novel therapeutic approaches as well as a deeper understanding of ADP-ribosylation as a whole.


Asunto(s)
ADP-Ribosilación/fisiología , Adenosina Difosfato/metabolismo , Hidrolasas/química , Hidrolasas/metabolismo , Humanos , Hidrolasas/clasificación , Dominios Proteicos , Relación Estructura-Actividad
11.
Genes Dev ; 34(5-6): 360-394, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32029455

RESUMEN

Oxidative and replication stress underlie genomic instability of cancer cells. Amplifying genomic instability through radiotherapy and chemotherapy has been a powerful but nonselective means of killing cancer cells. Precision medicine has revolutionized cancer therapy by putting forth the concept of selective targeting of cancer cells. Poly(ADP-ribose) polymerase (PARP) inhibitors represent a successful example of precision medicine as the first drugs targeting DNA damage response to have entered the clinic. PARP inhibitors act through synthetic lethality with mutations in DNA repair genes and were approved for the treatment of BRCA mutated ovarian and breast cancer. PARP inhibitors destabilize replication forks through PARP DNA entrapment and induce cell death through replication stress-induced mitotic catastrophe. Inhibitors of poly(ADP-ribose) glycohydrolase (PARG) exploit and exacerbate replication deficiencies of cancer cells and may complement PARP inhibitors in targeting a broad range of cancer types with different sources of genomic instability. Here I provide an overview of the molecular mechanisms and cellular consequences of PARP and PARG inhibition. I highlight clinical performance of four PARP inhibitors used in cancer therapy (olaparib, rucaparib, niraparib, and talazoparib) and discuss the predictive biomarkers of inhibitor sensitivity, mechanisms of resistance as well as the means of overcoming them through combination therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Glicósido Hidrolasas/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Inestabilidad Genómica , Glicósido Hidrolasas/metabolismo , Humanos , Neoplasias/enzimología , Poli(ADP-Ribosa) Polimerasas/metabolismo
12.
Genes Dev ; 34(5-6): 302-320, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32029452

RESUMEN

ADP-ribosylation (ADPRylation) is a posttranslational modification of proteins discovered nearly six decades ago, but many important questions remain regarding its molecular functions and biological roles, as well as the activity of the ADP-ribose (ADPR) transferase enzymes (PARP family members) that catalyze it. Growing evidence indicates that PARP-mediated ADPRylation events are key regulators of the protein biosynthetic pathway, leading from rDNA transcription and ribosome biogenesis to mRNA synthesis, processing, and translation. In this review we describe the role of PARP proteins and ADPRylation in all facets of this pathway. PARP-1 and its enzymatic activity are key regulators of rDNA transcription, which is a critical step in ribosome biogenesis. An emerging role of PARPs in alternative splicing of mRNAs, as well as direct ADPRylation of mRNAs, highlight the role of PARP members in RNA processing. Furthermore, PARP activity, stimulated by cellular stresses, such as viral infections and ER stress, leads to the regulation of mRNA stability and protein synthesis through posttranscriptional mechanisms. Dysregulation of PARP activity in these processes can promote disease states. Collectively, these results highlight the importance of PARP family members and ADPRylation in gene regulation, mRNA processing, and protein abundance. Future studies in these areas will yield new insights into the fundamental mechanisms and a broader utility for PARP-targeted therapeutic agents.


Asunto(s)
ADP-Ribosilación/fisiología , Expresión Génica/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Biosíntesis de Proteínas/fisiología , Proteostasis/fisiología , Animales , Humanos , Procesamiento Proteico-Postraduccional , ARN/metabolismo
13.
Mol Cell ; 73(4): 845-856.e5, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30712989

RESUMEN

ADP-ribosylation refers to the addition of one or more ADP-ribose groups onto proteins. The attached ADP-ribose monomers or polymers, commonly known as poly(ADP-ribose) (PAR), modulate the activities of the modified substrates or their binding affinities to other proteins. However, progress in this area is hindered by a lack of tools to investigate this protein modification. Here, we describe a new method named ELTA (enzymatic labeling of terminal ADP-ribose) for labeling free or protein-conjugated ADP-ribose monomers and polymers at their 2'-OH termini using the enzyme OAS1 and dATP. When coupled with various dATP analogs (e.g., radioactive, fluorescent, affinity tags), ELTA can be used to explore PAR biology with techniques routinely used to investigate DNA or RNA function. We demonstrate that ELTA enables the biophysical measurements of protein binding to PAR of a defined length, detection of PAR length from proteins and cells, and enrichment of sub-femtomole amounts of ADP-ribosylated peptides from cell lysates.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/metabolismo , ADP-Ribosilación , Adenosina Difosfato Ribosa/metabolismo , Nucleótidos de Desoxiadenina/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , Animales , Células HeLa , Humanos , Unión Proteica , Dominios Proteicos , Células Sf9 , Ubiquitina-Proteína Ligasas/genética
14.
Mol Cell ; 75(6): 1286-1298.e12, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31473101

RESUMEN

Long interspersed element-1 (LINE-1 or L1) retrotransposition poses a threat to genome integrity, and cells have evolved mechanisms to restrict retrotransposition. However, how cellular proteins facilitate L1 retrotransposition requires elucidation. Here, we demonstrate that single-strand DNA breaks induced by the L1 endonuclease trigger the recruitment of poly(ADP-ribose) polymerase 2 (PARP2) to L1 integration sites and that PARP2 activation leads to the subsequent recruitment of the replication protein A (RPA) complex to facilitate retrotransposition. We further demonstrate that RPA directly binds activated PARP2 through poly(ADP-ribosyl)ation and can protect single-strand L1 integration intermediates from APOBEC3-mediated cytidine deamination in vitro. Paradoxically, we provide evidence that RPA can guide APOBEC3A, and perhaps other APOBEC3 proteins, to sites of L1 integration. Thus, the interplay of L1-encoded and evolutionarily conserved cellular proteins is required for efficient retrotransposition; however, these interactions also may be exploited to restrict L1 retrotransposition in the human genome.


Asunto(s)
Elementos de Nucleótido Esparcido Largo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteína de Replicación A/metabolismo , Desaminasas APOBEC , Animales , Células CHO , Cricetulus , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Células HEK293 , Células HeLa , Humanos , Poli(ADP-Ribosa) Polimerasas/genética , Unión Proteica , Proteína de Replicación A/genética
15.
Mol Cell ; 75(6): 1270-1285.e14, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31351877

RESUMEN

PARP inhibitors (PARPi) prevent cancer cell growth by inducing synthetic lethality with DNA repair defects (e.g., in BRCA1/2 mutant cells). We have identified an alternative pathway for PARPi-mediated growth control in BRCA1/2-intact breast cancer cells involving rDNA transcription and ribosome biogenesis. PARP-1 binds to snoRNAs, which stimulate PARP-1 catalytic activity in the nucleolus independent of DNA damage. Activated PARP-1 ADP-ribosylates DDX21, an RNA helicase that localizes to nucleoli and promotes rDNA transcription when ADP-ribosylated. Treatment with PARPi or mutation of the ADP-ribosylation sites reduces DDX21 nucleolar localization, rDNA transcription, ribosome biogenesis, protein translation, and cell growth. The salient features of this pathway are evident in xenografts in mice and human breast cancer patient samples. Elevated levels of PARP-1 and nucleolar DDX21 are associated with cancer-related outcomes. Our studies provide a mechanistic rationale for efficacy of PARPi in cancer cells lacking defects in DNA repair whose growth is inhibited by PARPi.


Asunto(s)
Neoplasias de la Mama/metabolismo , ARN Helicasas DEAD-box/metabolismo , Proteínas de Neoplasias/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , ARN Neoplásico/metabolismo , ARN Nucleolar Pequeño/metabolismo , Ribosomas/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , ARN Helicasas DEAD-box/genética , Reparación del ADN , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Neoplasias/genética , Poli(ADP-Ribosa) Polimerasa-1/genética , ARN Neoplásico/genética , ARN Nucleolar Pequeño/genética , Ribosomas/genética
16.
Trends Biochem Sci ; 47(5): 390-402, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34366182

RESUMEN

Poly-ADP-ribose-polymerases (PARPs) are a family of 17 enzymes that regulate a diverse range of cellular processes in mammalian cells. PARPs catalyze the transfer of ADP-ribose from NAD+ to target molecules, most prominently amino acids on protein substrates, in a process known as ADP-ribosylation. Identifying the direct protein substrates of individual PARP family members is an essential first step for elucidating the mechanism by which PARPs regulate a particular pathway in cells. Two distinct chemical genetic (CG) strategies have been developed for identifying the direct protein substrates of individual PARP family members. In this review, we discuss the design principles behind these two strategies and how target identification has provided novel insight into the cellular function of individual PARPs and PARP-mediated ADP-ribosylation.


Asunto(s)
ADP-Ribosilación , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Adenosina Difosfato Ribosa/metabolismo , Animales , Mamíferos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Poli(ADP-Ribosa) Polimerasas/química , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas/metabolismo
17.
Mol Cell ; 71(5): 703-717.e9, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30100264

RESUMEN

In amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD), cytoplasmic aggregates of hyperphosphorylated TDP-43 accumulate and colocalize with some stress granule components, but how pathological TDP-43 aggregation is nucleated remains unknown. In Drosophila, we establish that downregulation of tankyrase, a poly(ADP-ribose) (PAR) polymerase, reduces TDP-43 accumulation in the cytoplasm and potently mitigates neurodegeneration. We establish that TDP-43 non-covalently binds to PAR via PAR-binding motifs embedded within its nuclear localization sequence. PAR binding promotes liquid-liquid phase separation of TDP-43 in vitro and is required for TDP-43 accumulation in stress granules in mammalian cells and neurons. Stress granule localization initially protects TDP-43 from disease-associated phosphorylation, but upon long-term stress, stress granules resolve, leaving behind aggregates of phosphorylated TDP-43. Finally, small-molecule inhibition of Tankyrase-1/2 in mammalian cells inhibits formation of cytoplasmic TDP-43 foci without affecting stress granule assembly. Thus, Tankyrase inhibition antagonizes TDP-43-associated pathology and neurodegeneration and could have therapeutic utility for ALS and FTD.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Células COS , Línea Celular , Núcleo Celular/metabolismo , Chlorocebus aethiops , Citoplasma/metabolismo , Drosophila , Femenino , Degeneración Lobar Frontotemporal/metabolismo , Masculino , Mamíferos/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley
18.
Mol Cell ; 72(1): 127-139.e8, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30244837

RESUMEN

The BRCA1 tumor suppressor preserves genome integrity through both homology-directed repair (HDR) and stalled fork protection (SFP). In vivo, BRCA1 exists as a heterodimer with the BARD1 tumor suppressor, and both proteins harbor a phosphate-binding BRCT domain. Here, we compare mice with mutations that ablate BRCT phospho-recognition by Bard1 (Bard1S563F and Bard1K607A) or Brca1 (Brca1S1598F). Brca1S1598F abrogates both HDR and SFP, suggesting that both pathways are likely impaired in most BRCA1 mutant tumors. Although not affecting HDR, the Bard1 mutations ablate poly(ADP-ribose)-dependent recruitment of BRCA1/BARD1 to stalled replication forks, resulting in fork degradation and chromosome instability. Nonetheless, Bard1S563F/S563F and Bard1K607A/K607A mice, unlike Brca1S1598F/S1598F mice, are not tumor prone, indicating that HDR alone is sufficient to suppress tumor formation in the absence of SFP. Nevertheless, because SFP, unlike HDR, is also impaired in heterozygous Brca1/Bard1 mutant cells, SFP and HDR may contribute to distinct stages of tumorigenesis in BRCA1/BARD1 mutation carriers.


Asunto(s)
Reparación del ADN/genética , Reparación del ADN por Recombinación/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Proteína BRCA1 , Inestabilidad Cromosómica/genética , Roturas del ADN de Doble Cadena , Femenino , Humanos , Ratones , Mutación , Dominios Proteicos/genética
19.
Proc Natl Acad Sci U S A ; 120(44): e2312999120, 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37883434

RESUMEN

Tpt1 is an essential agent of fungal and plant tRNA splicing that removes an internal RNA 2'-phosphate generated by tRNA ligase. Tpt1 also removes the 2'-phosphouridine mark installed by Ark1 kinase in the V-loop of archaeal tRNAs. Tpt1 performs a two-step reaction in which the 2'-PO4 attacks NAD+ to form an RNA-2'-phospho-(ADP-ribose) intermediate, and transesterification of the ADP-ribose O2″ to the RNA 2'-phosphodiester yields 2'-OH RNA and ADP-ribose-1″,2″-cyclic phosphate. Here, we present structures of archaeal Tpt1 enzymes, captured as product complexes with ADP-ribose-1″-PO4, ADP-ribose-2″-PO4, and 2'-OH RNA, and as substrate complexes with 2',5'-ADP and NAD+, that illuminate 2'-PO4 junction recognition and catalysis. We show that archaeal Tpt1 enzymes can use the 2'-PO4-containing metabolites NADP+ and NADPH as substrates for 2'-PO4 transfer to NAD+. A role in 2'-phospho-NADP(H) dynamics provides a rationale for the prevalence of Tpt1 in taxa that lack a capacity for internal RNA 2'-phosphorylation.


Asunto(s)
NAD , ARN , ARN/metabolismo , NADP , NAD/metabolismo , ARN de Transferencia/genética , Adenosina Difosfato Ribosa/metabolismo , Fosfatos/metabolismo
20.
Proc Natl Acad Sci U S A ; 120(19): e2215068120, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37126687

RESUMEN

Poly(ADP-ribose) (PAR) is a homopolymer of adenosine diphosphate ribose that is added to proteins as a posttranslational modification to regulate numerous cellular processes. PAR also serves as a scaffold for protein binding in macromolecular complexes, including biomolecular condensates. It remains unclear how PAR achieves specific molecular recognition. Here, we use single-molecule fluorescence resonance energy transfer (smFRET) to evaluate PAR flexibility under different cation conditions. We demonstrate that, compared to RNA and DNA, PAR has a longer persistence length and undergoes a sharper transition from extended to compact states in physiologically relevant concentrations of various cations (Na+, Mg2+, Ca2+, and spermine4+). We show that the degree of PAR compaction depends on the concentration and valency of cations. Furthermore, the intrinsically disordered protein FUS also served as a macromolecular cation to compact PAR. Taken together, our study reveals the inherent stiffness of PAR molecules, which undergo switch-like compaction in response to cation binding. This study indicates that a cationic environment may drive recognition specificity of PAR.


Asunto(s)
Adenosina Difosfato Ribosa , Poli Adenosina Difosfato Ribosa , Poli Adenosina Difosfato Ribosa/química , Poli Adenosina Difosfato Ribosa/metabolismo , Adenosina Difosfato Ribosa/química , Procesamiento Proteico-Postraduccional , Unión Proteica , Fenómenos Fisiológicos Celulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA