Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Mol Cell ; 63(3): 485-97, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27425408

RESUMO

Pro-apoptotic BAX is a cell fate regulator playing an important role in cellular homeostasis and pathological cell death. BAX is predominantly localized in the cytosol, where it has a quiescent monomer conformation. Following a pro-apoptotic trigger, cytosolic BAX is activated and translocates to the mitochondria to initiate mitochondrial dysfunction and apoptosis. Here, cellular, biochemical, and structural data unexpectedly demonstrate that cytosolic BAX also has an inactive dimer conformation that regulates its activation. The full-length crystal structure of the inactive BAX dimer revealed an asymmetric interaction consistent with inhibition of the N-terminal conformational change of one protomer and the displacement of the C-terminal helix α9 of the second protomer. This autoinhibited BAX dimer dissociates to BAX monomers before BAX can be activated. Our data support a model whereby the degree of apoptosis induction is regulated by the conformation of cytosolic BAX and identify an unprecedented mechanism of cytosolic BAX inhibition.


Assuntos
Apoptose , Transdução de Sinais , Proteína X Associada a bcl-2/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Cristalografia por Raios X , Citosol/metabolismo , Fibroblastos/metabolismo , Humanos , Camundongos , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Relação Estrutura-Atividade , Transfecção , Proteína X Associada a bcl-2/química , Proteína X Associada a bcl-2/genética
2.
Mol Cell ; 57(5): 873-886, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25684204

RESUMO

BCL-2 is a negative regulator of apoptosis implicated in homeostatic and pathologic cell survival. The canonical anti-apoptotic mechanism involves entrapment of activated BAX by a groove on BCL-2, preventing BAX homo-oligomerization and mitochondrial membrane poration. The BCL-2 BH4 domain also confers anti-apoptotic functionality, but the mechanism is unknown. We find that a synthetic α-helical BH4 domain binds to BAX with nanomolar affinity and independently inhibits the conformational activation of BAX. Hydrogen-deuterium exchange mass spectrometry demonstrated that the N-terminal conformational changes in BAX induced by a triggering BIM BH3 helix were suppressed by the BCL-2 BH4 helix. Structural analyses localized the BH4 interaction site to a groove formed by residues of α1, α1-α2 loop, and α2-α3 and α5-α6 hairpins on the BAX surface. These data reveal a previously unappreciated binding site for targeted inhibition of BAX and suggest that the BCL-2 BH4 domain may participate in apoptosis blockade by a noncanonical interaction mechanism.


Assuntos
Apoptose , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/química , Proteína X Associada a bcl-2/química , Sequência de Aminoácidos , Sítios de Ligação/genética , Medição da Troca de Deutério/métodos , Células HeLa , Humanos , Espectrometria de Massas/métodos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Conformação Proteica , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
3.
Nat Chem Biol ; 15(4): 322-330, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30718816

RESUMO

BAX is a critical effector of the mitochondrial cell death pathway in response to a diverse range of stimuli in physiological and disease contexts. Upon binding by BH3-only proteins, cytosolic BAX undergoes conformational activation and translocation, resulting in mitochondrial outer-membrane permeabilization. Efforts to rationally target BAX and develop inhibitors have been elusive, despite the clear therapeutic potential of inhibiting BAX-mediated cell death in a host of diseases. Here, we describe a class of small-molecule BAX inhibitors, termed BAIs, that bind directly to a previously unrecognized pocket and allosterically inhibit BAX activation. BAI binding around the hydrophobic helix α5 using hydrophobic and hydrogen bonding interactions stabilizes key areas of the hydrophobic core. BAIs inhibit conformational events in BAX activation that prevent BAX mitochondrial translocation and oligomerization. Our data highlight a novel paradigm for effective and selective pharmacological targeting of BAX to enable rational development of inhibitors of BAX-mediated cell death.


Assuntos
Proteína X Associada a bcl-2/antagonistas & inibidores , Proteína X Associada a bcl-2/metabolismo , Sequência de Aminoácidos , Apoptose/fisiologia , Sítios de Ligação/fisiologia , Cromatografia Gasosa-Espectrometria de Massas/métodos , Humanos , Mitocôndrias/fisiologia , Modelos Moleculares , Fragmentos de Peptídeos/fisiologia , Permeabilidade , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
5.
Nat Chem Biol ; 9(6): 374-82, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23584676

RESUMO

Chaperone-mediated autophagy (CMA) contributes to cellular quality control and the cellular response to stress through the selective degradation of cytosolic proteins in lysosomes. A decrease in CMA activity occurs in aging and in age-related disorders (for example, neurodegenerative diseases and diabetes). Although prevention of this age-dependent decline through genetic manipulation in mice has proven beneficial, chemical modulation of CMA is not currently possible, owing in part to the lack of information on the signaling mechanisms that modulate this pathway. In this work, we report that signaling through retinoic acid receptor α (RARα) inhibits CMA and apply structure-based chemical design to develop synthetic derivatives of all-trans-retinoic acid to specifically neutralize this inhibitory effect. We demonstrate that chemical enhancement of CMA protects cells from oxidative stress and from proteotoxicity, supporting a potential therapeutic opportunity when reduced CMA contributes to cellular dysfunction and disease.


Assuntos
Autofagia , Chaperonas Moleculares/química , Tretinoína/química , Animais , Sítios de Ligação , Citosol/metabolismo , DNA/química , Lisossomos/metabolismo , Camundongos , Conformação Molecular , Simulação de Dinâmica Molecular , Células NIH 3T3 , Oxigênio/metabolismo , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico
6.
Mar Drugs ; 12(8): 4311-25, 2014 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-25076060

RESUMO

A series of novel marinopyrroles with sulfide and sulphone spacers were designed and synthesized. Their activity to disrupt the binding of the pro-apoptotic protein, Bim, to the pro-survival proteins, Mcl-1 and Bcl-xL, was evaluated using ELISA assays. Fluorescence-quenching (FQ) assays confirmed the direct binding of marinopyrroles to Mcl-1. Benzyl- and benzyl methoxy-containing sulfide derivatives 4 and 5 were highly potent dual Mcl-1/Bim and Bcl-xL/Bim disruptors (IC50 values of 600 and 700 nM), whereas carboxylate-containing sulfide derivative 9 exhibited 16.4-fold more selectivity for disrupting Mcl-1/Bim over Bcl-xL/Bim binding. In addition, a nonsymmetrical marinopyrrole 12 is as equally potent as the parent marinopyrrole A (1) for disrupting both Mcl-1/Bim and Bcl-xL/Bim binding. Some of the derivatives were also active in intact human breast cancer cells where they reduced the levels of Mcl-1, induced programd cell death (apoptosis) and inhibited cell proliferation.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Membrana/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Pirróis/farmacologia , Sulfetos/farmacologia , Apoptose/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2 , Linhagem Celular Tumoral , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo
7.
Proc Natl Acad Sci U S A ; 108(40): 16600-5, 2011 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-21949356

RESUMO

Mutations in rumi result in a temperature-sensitive loss of Notch signaling in Drosophila. Drosophila Rumi is a soluble, endoplasmic reticulum-retained protein with a CAP10 domain that functions as a protein O-glucosyltransferase. In human and mouse genomes, three potential Rumi homologues exist: one with a high degree of identity to Drosophila Rumi (52%), and two others with lower degrees of identity but including a CAP10 domain (KDELC1 and KDELC2). Here we show that both mouse and human Rumi, but not KDELC1 or KDELC2, catalyze transfer of glucose from UDP-glucose to an EGF repeat from human factor VII. Similarly, human Rumi, but not KDELC1 or KDELC2, rescues the Notch phenotypes in Drosophila rumi clones. During characterization of the Rumi enzymes, we noted that, in addition to protein O-glucosyltransferase activity, both mammalian and Drosophila Rumi also showed significant protein O-xylosyltransferase activity. Rumi transfers Xyl or glucose to serine 52 in the O-glucose consensus sequence ( ) of factor VII EGF repeat. Surprisingly, the second serine (S53) facilitates transfer of Xyl, but not glucose, to the EGF repeat by Rumi. EGF16 of mouse Notch2, which has a diserine motif in the consensus sequence ( ), is also modified with either O-Xyl or O-glucose glycans in cells. Mutation of the second serine (S590A) causes a loss of O-Xyl but not O-glucose at this site. Altogether, our data establish dual substrate specificity for the glycosyltransferase Rumi and provide evidence that amino acid sequences of the recipient EGF repeat significantly influence which donor substrate (UDP-glucose or UDP-Xyl) is used.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Fator VII/metabolismo , Glucosiltransferases/metabolismo , Pentosiltransferases/metabolismo , Transdução de Sinais/fisiologia , Animais , Drosophila/genética , Proteínas de Drosophila/genética , Fator VII/genética , Glucosiltransferases/genética , Humanos , Espectrometria de Massas , Camundongos , Mutação/genética , Transdução de Sinais/genética , Especificidade por Substrato , UDP Xilose-Proteína Xilosiltransferase
8.
J Proteome Res ; 11(3): 1969-80, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22268864

RESUMO

The diverse influences of ubiquitin, mediated by its post-translational covalent modification of other proteins, have been extensively investigated. However, more recently roles for unanchored (nonsubstrate linked) polyubiquitin chains have also been proposed. Here we describe the use of ubiquitin-binding domains to affinity purify endogenous unanchored polyubiquitin chains and their subsequent characterization by mass spectrometry (MS). Using the A20 Znf domain of the ubiquitin receptor ZNF216 we isolated a protein from skeletal muscle shown by a combination of nanoLC-MS and LC-MS/MS to represent an unmodified and unanchored K48-linked ubiquitin dimer. Selective purification of unanchored polyubiquitin chains using the Znf UBP (BUZ) domain of USP5/isopeptidase-T allowed the isolation of K48 and K11-linked ubiquitin dimers, as well as revealing longer chains containing as many as 15 ubiquitin moieties, which include the K48 linkage. Top-down nanoLC-MS/MS of the A20 Znf-purified ubiquitin dimer generated diagnostic ions consistent with the presence of the K48 linkage, illustrating for the first time the potential of this approach to probe connectivity within endogenous polyubiquitin modifications. As well as providing initial proteomic insights into the molecular composition of endogenous unanchored polyubiquitin chains, this work also represents the first definition of polyubiquitin chain length in vivo.


Assuntos
Poliubiquitina/metabolismo , Proteínas Ubiquitinadas/metabolismo , Sequência de Aminoácidos , Animais , Cromatografia de Afinidade/métodos , Proteínas de Ligação a DNA/química , Humanos , Proteínas Imobilizadas/química , Masculino , Músculo Esquelético/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/isolamento & purificação , Poliubiquitina/química , Poliubiquitina/isolamento & purificação , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Espectrometria de Massas em Tandem , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Proteínas Ubiquitinadas/química , Proteínas Ubiquitinadas/isolamento & purificação
9.
Biochem Soc Trans ; 40(2): 404-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22435820

RESUMO

UBDs [Ub (ubiquitin)-binding domains], which are typically small protein motifs of <50 residues, are used by receptor proteins to transduce post-translational Ub modifications in a wide range of biological processes, including NF-κB (nuclear factor κB) signalling and proteasomal degradation pathways. More than 20 families of UBDs have now been characterized in structural detail and, although many recognize the canonical Ile44/Val70-binding patch on Ub, a smaller number have alternative Ub-recognition sites. The A20 Znf (A20-like zinc finger) of the ZNF216 protein is one of the latter and binds with high affinity to a polar site on Ub centred around Asp58/Gln62. ZNF216 shares some biological function with p62, with both linked to NF-κB signal activation and as shuttle proteins in proteasomal degradation pathways. The UBA domain (Ub-associated domain) of p62, although binding to Ub through the Ile44/Val70 patch, is unique in forming a stable dimer that negatively regulates Ub recognition. We show that the A20 Znf and UBA domain are able to form a ternary complex through independent interactions with a single Ub molecule, supporting functional models for Ub as a 'hub' for mediating multi-protein complex assembly and for enhancing signalling specificity.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Ubiquitina/metabolismo , Animais , Humanos , Mutação/genética , Osteíte Deformante/genética , Ligação Proteica , Estrutura Terciária de Proteína
10.
Nat Commun ; 13(1): 4220, 2022 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-35864098

RESUMO

Chaperone-mediated autophagy activity, essential in the cellular defense against proteotoxicity, declines with age, and preventing this decline in experimental genetic models has proven beneficial. Here, we have identified the mechanism of action of selective chaperone-mediated autophagy activators previously developed by our group and have leveraged that information to generate orally bioavailable chaperone-mediated autophagy activators with favorable brain exposure. Chaperone-mediated autophagy activating molecules stabilize the interaction between retinoic acid receptor alpha - a known endogenous inhibitor of chaperone-mediated autophagy - and its co-repressor, nuclear receptor corepressor 1, resulting in changes of a discrete subset of the retinoic acid receptor alpha transcriptional program that leads to selective chaperone-mediated autophagy activation. Chaperone-mediated autophagy activators molecules activate this pathway in vivo and ameliorate retinal degeneration in a retinitis pigmentosa mouse model. Our findings reveal a mechanism for pharmacological targeting of chaperone-mediated autophagy activation and suggest a therapeutic strategy for retinal degeneration.


Assuntos
Autofagia Mediada por Chaperonas , Degeneração Retiniana , Retinose Pigmentar , Animais , Autofagia , Proteínas Correpressoras , Camundongos , Receptor alfa de Ácido Retinoico/genética
11.
Nat Commun ; 13(1): 3775, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35798717

RESUMO

Mitofusins reside on the outer mitochondrial membrane and regulate mitochondrial fusion, a physiological process that impacts diverse cellular processes. Mitofusins are activated by conformational changes and subsequently oligomerize to enable mitochondrial fusion. Here, we identify small molecules that directly increase or inhibit mitofusins activity by modulating mitofusin conformations and oligomerization. We use these small molecules to better understand the role of mitofusins activity in mitochondrial fusion, function, and signaling. We find that mitofusin activation increases, whereas mitofusin inhibition decreases mitochondrial fusion and functionality. Remarkably, mitofusin inhibition also induces minority mitochondrial outer membrane permeabilization followed by sub-lethal caspase-3/7 activation, which in turn induces DNA damage and upregulates DNA damage response genes. In this context, apoptotic death induced by a second mitochondria-derived activator of caspases (SMAC) mimetic is potentiated by mitofusin inhibition. These data provide mechanistic insights into the function and regulation of mitofusins as well as small molecules to pharmacologically target mitofusins.


Assuntos
GTP Fosfo-Hidrolases , Mitocôndrias , GTP Fosfo-Hidrolases/metabolismo , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Transdução de Sinais
12.
Biochemistry ; 50(21): 4665-74, 2011 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-21517082

RESUMO

The scaffold protein p62/SQSTM1 acts as a hub in regulating a diverse range of signaling pathways which are dependent upon a functional ubiquitin-binding C-terminal UBA domain. Mutations linked to Paget's disease of bone (PDB) commonly cluster within the UBA domain. The p62 UBA domain is unique in forming a highly stable dimer which regulates ubiquitin recognition by using overlapping surface patches in both dimerization and ubiquitin binding, making the two association events competitive. NMR structural analysis and biophysical methods show that some PDB mutations modulated the ubiquitin binding affinity by both direct and indirect mechanisms that affect UBA structural integrity, dimer stability, and contacts at the UBA-ubiquitin interface. In other cases, common PDB mutations (P392L in particular) result in no significant change in ubiquitin binding affinity for the UBA domain in isolation; however, all PDB UBA mutations lead to loss of function with respect to ubiquitin binding in the context of full-length p62, suggesting a more complex underlying mechanism.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Mutação , Osteíte Deformante/genética , Ubiquitina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Dimerização , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Proteína Sequestossoma-1
13.
Biochemistry ; 50(42): 9076-87, 2011 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-21923101

RESUMO

Ubiquitin (Ub) modifications are transduced by receptor proteins that use Ub-binding domains (UBDs) to recognize distinct interaction faces on the Ub surface. We report the nuclear magnetic resonance (NMR) solution structures of the A20-like zinc finger (A20 Znf) UBD of the Ub receptor ZNF216, and its complex with Ub, and show that the binding surface on Ub centered on Asp58 leaves the canonical hydrophobic Ile44 patch free to participate in additional interactions. We have modeled ternary complexes of the different families of UBDs and show that while many are expected to bind competitively to the same Ile44 surface or show steric incompatibility, other combinations (in particular, those involving the A20 Znf domain) are consistent with a single Ub moiety simultaneously participating in multiple interactions with different UBDs. We subsequently demonstrate by NMR that the A20 Znf domain of ZNF216 and the UBA domain of the p62 protein (an Ile44-binding UBD), which function in the same biological pathways, are able to form such a Ub-mediated ternary complex through independent interactions with a single Ub. This work supports an emerging concept of Ub acting as a scaffold to mediate multiprotein complex assembly.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Dedos de Zinco , Motivos de Aminoácidos/genética , Animais , Ácido Aspártico/metabolismo , Linhagem Celular Tumoral , Cristalografia por Raios X , Proteínas de Ligação a DNA/genética , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Proteínas Musculares/química , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutagênese Sítio-Dirigida , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Ratos , Transdução de Sinais/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Ubiquitina-Proteína Ligases/genética , Dedos de Zinco/genética
14.
J Am Chem Soc ; 133(4): 1044-51, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21162526

RESUMO

The structurally unique natural product telomestatin incorporates seven oxazole rings and one sulfur-containing thiazoline in a macrocyclic arrangement. The compound is a potent inhibitor of the enzyme telomerase and therefore provides a structural framework for developing new potential therapeutic agents for cancer. An efficient formal total synthesis of telomestatin is reported in which the key steps are the use of dirhodium(II)-catalyzed reactions of diazocarbonyl compounds to generate six oxazole rings, demonstrating the power of rhodium carbene methodology in organic chemical synthesis. CD spectroscopy establishes that seco-derivatives of telomestatin are potent stabilizers of G-quadruplex structures derived from the human telomeric repeat sequence. Mass spectrometry studies, confirmed by molecular dynamics simulations, provide the first evidence that high affinity binding to terminal G-tetrads in both 1:1 and 2:1 ligand complexes is mediated through the macrocycle coordinating a monovalent cation, with selectivity for the antiparallel structure.


Assuntos
Quadruplex G/efeitos dos fármacos , Oxazóis/síntese química , Oxazóis/farmacologia , Sequência de Bases , Fenômenos Biofísicos , Cátions Monovalentes/química , Humanos , Ligantes , Metano/análogos & derivados , Metano/química , Modelos Moleculares , Oxazóis/química , Ródio/química
15.
Nat Commun ; 12(1): 1134, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33602934

RESUMO

The BCL-2 family protein BAX has essential activity in mitochondrial regulation of cell death. While BAX activity ensures tissue homeostasis, when dysregulated it contributes to aberrant cell death in several diseases. During cellular stress BAX is transformed from an inactive cytosolic conformation to a toxic mitochondrial oligomer. Although the BAX transformation process is not well understood, drugs that interfere with this process are useful research tools and potential therapeutics. Here, we show that Eltrombopag,  an FDA-approved drug,  is a direct inhibitor of BAX. Eltrombopag binds the BAX trigger site distinctly from BAX activators, preventing them from triggering BAX conformational transformation and simultaneously promoting stabilization of the inactive BAX structure. Accordingly, Eltrombopag is capable of inhibiting BAX-mediated apoptosis induced by cytotoxic stimuli. Our data demonstrate structure-function insights into a mechanism of BAX inhibition and reveal a mechanism for Eltrombopag that may expand its use in diseases of uncontrolled cell death.


Assuntos
Benzoatos/farmacologia , Hidrazinas/farmacologia , Pirazóis/farmacologia , Proteína X Associada a bcl-2/antagonistas & inibidores , Células 3T3 , Animais , Apoptose/efeitos dos fármacos , Benzoatos/química , Morte Celular/efeitos dos fármacos , Humanos , Hidrazinas/química , Espectroscopia de Ressonância Magnética , Camundongos , Modelos Biológicos , Modelos Moleculares , Estabilidade Proteica/efeitos dos fármacos , Pirazóis/química , Proteína X Associada a bcl-2/química , Proteína X Associada a bcl-2/metabolismo
16.
Biochemistry ; 49(35): 7625-33, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20715834

RESUMO

The 5' end of exon C of the bovine estrogen receptor alpha gene (bov-ESR1) includes a unique G-rich insert, not found in other closely related mammalian genes, which lies close to both a double E-box transcription factor binding site and the site of a single nucleotide (G/A) polymorphism. Biophysical studies, using CD and UV absorbance measurements, show that this 22 base insert leads to the formation of a family of stable G-quadruplex structures which are unaffected by the G/A polymorphism. Multiplex PCR shows that the region including the G-quadruplex is transcribed into RNA, and studies with a synthetic RNA transcript sequence demonstrated formation of a highly stable parallel-folded quadruplex structure. Luciferase reporter constructs demonstrate that the G-rich sequence reduces rates of translation when present in the 5'-UTR of mRNA transcripts. Mutations (GGG to AAA) that destabilize the quadruplex lead to a 15-fold enhancement of translational efficiency, suggesting that a possible biological role of the insert in exon C of the bov-ESR1 is to regulate translation of this exon.


Assuntos
Receptor alfa de Estrogênio/genética , Éxons , Quadruplex G , Regiões 5' não Traduzidas , Animais , Sequência de Bases , Bovinos , Dicroísmo Circular , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA Mensageiro/genética
17.
Nat Cancer ; 1(3): 315-328, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32776015

RESUMO

Doxorubicin remains an essential component of many cancer regimens, but its use is limited by lethal cardiomyopathy, which has been difficult to target, owing to pleiotropic mechanisms leading to apoptotic and necrotic cardiac cell death. Here we show that BAX is rate-limiting in doxorubicin-induced cardiomyopathy and identify a small-molecule BAX inhibitor that blocks both apoptosis and necrosis to prevent this syndrome. By allosterically inhibiting BAX conformational activation, this compound blocks BAX translocation to mitochondria, thereby abrogating both forms of cell death. When co-administered with doxorubicin, this BAX inhibitor prevents cardiomyopathy in zebrafish and mice. Notably, cardioprotection does not compromise the efficacy of doxorubicin in reducing leukemia or breast cancer burden in vivo, primarily due to increased priming of mitochondrial death mechanisms and higher BAX levels in cancer cells. This study identifies BAX as an actionable target for doxorubicin-induced cardiomyopathy and provides a prototype small-molecule therapeutic.


Assuntos
Cardiomiopatias , Peixe-Zebra , Animais , Apoptose/fisiologia , Cardiomiopatias/induzido quimicamente , Doxorrubicina/efeitos adversos , Camundongos , Necrose , Peixe-Zebra/metabolismo , Proteína X Associada a bcl-2
18.
Biochemistry ; 48(48): 11487-95, 2009 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-19860473

RESUMO

Tissue-specific expression of the human estrogen receptor alpha gene (ESR1) is achieved through multiple promoter sequences resulting in various mRNA transcripts encoding a common protein but differing in their 5'-untranslated region (5'-UTR). Many cancers are estrogen-sensitive with neoplastic growth stimulated through the estrogen receptor, a transcription factor that regulates developmental genes. We demonstrate that the human ESR1 gene is rich in potential quadruplex-forming sequences with 3 of 20 identified within exonic regions. In particular, we show using CD, UV, and NMR spectroscopy that a stable DNA G-quadruplex motif is formed within the exon C gene sequence. This motif, which PCR shows is transcribed in normal and neoplastic endometrium and in MCF-7 cells, forms a stable RNA quadruplex demonstrable by CD and UV analysis. Cloning the exon C G-quadruplex sequence upstream of a luciferase reporter gene caused a 6-fold reduction of enzymatic activity compared to a mutant sequence. We conclude that the exon C G-quadruplex motif is present in the 5'-UTR of the mRNA transcript, where it modulates the efficiency of translation.


Assuntos
Receptor alfa de Estrogênio/biossíntese , Receptor alfa de Estrogênio/genética , Quadruplex G , Biossíntese de Proteínas , Regiões 5' não Traduzidas , Sequência de Bases , Neoplasias da Mama/química , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Dicroísmo Circular , Ativação Enzimática , Feminino , Humanos , Espectroscopia de Ressonância Magnética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espectrofotometria Ultravioleta
19.
Org Biomol Chem ; 7(20): 4194-200, 2009 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-19795057

RESUMO

We report CD, ESI-MS and molecular modelling studies of ligand binding interactions with DNA quadruplex structures derived from the human telomeric repeat sequence (h-Tel) and the proto-oncogenic c-kit promoter sequence. These sequences form anti-parallel (both 2 + 2 and 3 + 1) and parallel conformations, respectively, and demonstrate distinctively different degrees of structural plasticity in binding ligands. With h-Tel, we show that an extended heteroaromatic 1,4-triazole (TRZ), designed to exploit pi-stacking interactions and groove-specific contacts, shows some selectivity for parallel folds, however, the polycyclic fluorinated acridinium cation (RHPS4), which is a similarly potent telomerase inhibitor, shows selectivity for anti-parallel conformations implicating favourable interactions with lateral and diagonal loops. In contrast, the unique c-kit parallel-stranded quadruplex shows none of the structural plasticity of h-Tel with either ligand. We show by quantitative ESI-MS analysis that both sequences are able to bind a ligand on either end of the quadruplex. In the case of h-Tel the two sites have similar affinities, however, in the case of the c-kit quadruplex the affinities of the two sites are different and ligand-dependent. We demonstrate that two different small molecule architectures result in significant differences in selectivity for parallel and anti-parallel quadruplex structures that may guide quadruplex targeted drug-design.


Assuntos
Acridinas/química , Acridinas/farmacologia , DNA/química , Quadruplex G/efeitos dos fármacos , Triazóis/química , Triazóis/farmacologia , Dicroísmo Circular , DNA/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Ligantes , Espectrometria de Massas , Simulação de Dinâmica Molecular , Proteínas Proto-Oncogênicas c-kit/genética , Especificidade por Substrato , Telomerase/antagonistas & inibidores , Telômero/genética
20.
Methods Mol Biol ; 1877: 217-231, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30536009

RESUMO

Biomolecular nuclear magnetic resonance (NMR) is a powerful and versatile method for studying both protein-protein interactions (PPIs) and protein-small molecule binding. NMR has been used extensively in the investigation of BCL-2 family proteins revealing the structure of key family members, identifying binding partners and interaction sites, and screening small molecule modulators. In this chapter we discuss the application of NMR to identify interaction sites and structure determination of protein-protein and protein-small molecule complexes using two examples.


Assuntos
Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sítios de Ligação/fisiologia , Linhagem Celular , Humanos , Ressonância Magnética Nuclear Biomolecular/métodos , Ligação Proteica/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , Bibliotecas de Moléculas Pequenas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA