Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Angew Chem Int Ed Engl ; 62(51): e202312517, 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-37924230

ABSTRACT

DNA repair protein RAD51 is a key player in the homologous recombination pathway. Upon DNA damage, RAD51 is transported into the nucleus by BRCA2, where it can repair DNA double-strand breaks. Due to the structural complexity and dynamics, researchers have not yet clarified the mechanistic details of every step of RAD51 recruitment and DNA repair. RAD51 possesses an intrinsic tendency to form oligomeric structures, which make it challenging to conduct biochemical and biophysical investigations. Here, for the first time, we report on the isolation and characterization of a human monomeric RAD51 recombinant form, obtained through a double mutation, which preserves the protein's integrity and functionality. We investigated different buffers to identify the most suitable condition needed to definitively stabilize the monomer. The monomer of human RAD51 provides the community with a unique biological tool for investigating RAD51-mediated homologous recombination, and paves the way for more reliable structural, mechanistic, and drug discovery studies.


Subject(s)
Homologous Recombination , Neoplasms , Rad51 Recombinase , Recombinant Proteins , Humans , DNA Damage , DNA Repair , Neoplasms/genetics , Rad51 Recombinase/chemistry , Rad51 Recombinase/genetics , Rad51 Recombinase/isolation & purification , Mutation , Protein Stability , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
2.
J Med Chem ; 66(14): 9797-9822, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37440686

ABSTRACT

In cystic fibrosis (CF), deletion of phenylalanine 508 (F508del) in the CF transmembrane conductance regulator (CFTR) is associated to misfolding and defective gating of the mutant channel. One of the most promising CF drug targets is the ubiquitin ligase RNF5, which promotes F508del-CFTR degradation. Recently, the first ever reported inhibitor of RNF5 was discovered, i.e., the 1,2,4-thiadiazol-5-ylidene inh-2. Here, we designed and synthesized a series of new analogues to explore the structure-activity relationships (SAR) of this class of compounds. SAR efforts ultimately led to compound 16, which showed a greater F508del-CFTR corrector activity than inh-2, good tolerability, and no toxic side effects. Analogue 16 increased the basal level of autophagy similar to what has been described with RNF5 silencing. Furthermore, co-treatment with 16 significantly improved the F508del-CFTR rescue induced by the triple combination elexacaftor/tezacaftor/ivacaftor in CFBE41o- cells. These findings validate the 1,2,4-thiadiazolylidene scaffold for the discovery of novel RNF5 inhibitors and provide evidence to pursue this unprecedented strategy for the treatment of CF.


Subject(s)
Cystic Fibrosis , Thiadiazoles , Humans , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Thiadiazoles/pharmacology , Thiadiazoles/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Structure-Activity Relationship , Aminophenols , Benzodioxoles/pharmacology , Mutation , DNA-Binding Proteins/metabolism
3.
Int J Mol Sci ; 24(8)2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37108703

ABSTRACT

Most kinase inhibitors are designed to bind to highly homologous ATP-binding sites, which leads to promiscuity and possible off-target effects. Allostery is an alternative approach to pursuing selectivity. However, allostery is difficult to exploit due to the wide variety of underlying mechanisms and the potential involvement of long-range conformational effects that are difficult to pinpoint. GSK-3ß is involved in several pathologies. This critical target has an ATP-binding site that is highly homologous with the orthosteric sites of other kinases. Unsurprisingly, there is also great similarity between the ATP-binding sites of GSK-3ß and its isomer, which is not redundant and thus would benefit from selective inhibition. Allostery would also allow for a moderate and tunable inhibition, which is ideal for GSK-3ß, because this target is involved in multiple pathways, some of which must be preserved. However, despite considerable research efforts, only one allosteric GSK-3ß inhibitor has reached the clinic. Moreover, unlike other kinases, there are no X-ray structures of GSK-3ß in complex with allosteric inhibitors in the PDB data bank. This review aims to summarize the state of the art in allosteric GSK-3ß inhibitor investigations, highlighting the aspects that make this target challenging for an allosteric approach.


Subject(s)
Adenosine Triphosphate , Protein Kinase Inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Protein Kinase Inhibitors/chemistry , Binding Sites , Adenosine Triphosphate/metabolism
4.
Cancers (Basel) ; 15(6)2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36980703

ABSTRACT

In recent years, the RAD52 protein has been highlighted as a mediator of many DNA repair mechanisms. While RAD52 was initially considered to be a non-essential auxiliary factor, its inhibition has more recently been demonstrated to be synthetically lethal in cancer cells bearing mutations and inactivation of specific intracellular pathways, such as homologous recombination. RAD52 is now recognized as a novel and critical pharmacological target. In this review, we comprehensively describe the available structural and functional information on RAD52. The review highlights the pathways in which RAD52 is involved and the approaches to RAD52 inhibition. We discuss the multifaceted role of this protein, which has a complex, dynamic, and functional 3D superstructural arrangement. This complexity reinforces the need to further investigate and characterize RAD52 to solve a challenging mechanistic puzzle and pave the way for a robust drug discovery campaign.

5.
ACS Med Chem Lett ; 13(8): 1262-1269, 2022 Aug 11.
Article in English | MEDLINE | ID: mdl-35978685

ABSTRACT

RAD51 is an ATP-dependent recombinase, recruited by BRCA2 to mediate DNA double-strand breaks repair through homologous recombination and represents an attractive cancer drug target. Herein, we applied for the first-time protein-templated dynamic combinatorial chemistry on RAD51 as a hit identification strategy. Upon design of N-acylhydrazone-based dynamic combinatorial libraries, RAD51 showed a clear templating effect, amplifying 19 N-acylhydrazones. Screening against the RAD51-BRCA2 protein-protein interaction via ELISA assay afforded 10 inhibitors in the micromolar range. Further 19F NMR experiments revealed that 7 could bind RAD51 and be displaced by BRC4, suggesting an interaction in the same binding pocket of BRCA2. These results proved not only that ptDCC could be successfully applied on full-length oligomeric RAD51, but also that it could address the need of alternative strategies toward the identification of small-molecule PPI inhibitors.

6.
Int J Mol Sci ; 23(7)2022 Mar 31.
Article in English | MEDLINE | ID: mdl-35409221

ABSTRACT

Glycogen synthase kinase 3 beta (GSK-3ß) is an evolutionarily conserved serine-threonine kinase dysregulated in numerous pathologies, such as Alzheimer's disease and cancer. Even though GSK-3ß is a validated pharmacological target most of its inhibitors have two main limitations: the lack of selectivity due to the high homology that characterizes the ATP binding site of most kinases, and the toxicity that emerges from GSK-3ß complete inhibition which translates into the impairment of the plethora of pathways GSK-3ß is involved in. Starting from a 1D 19F NMR fragment screening, we set up several biophysical assays for the identification of GSK-3ß inhibitors capable of binding protein hotspots other than the ATP binding pocket or to the ATP binding pocket, but with an affinity able of competing with a reference binder. A phosphorylation activity assay on a panel of several kinases provided selectivity data that were further rationalized and corroborated by structural information on GSK-3ß in complex with the hit compounds. In this study, we identified promising fragments, inhibitors of GSK-3ß, while proposing an alternative screening workflow that allows facing the flaws that characterize the most common GSK-3ß inhibitors through the identification of selective inhibitors and/or inhibitors able to modulate GSK-3ß activity without leading to its complete inhibition.


Subject(s)
Alzheimer Disease , Adenosine Triphosphate/metabolism , Alzheimer Disease/metabolism , Binding Sites , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Phosphorylation
7.
J Exp Clin Cancer Res ; 41(1): 53, 2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35135603

ABSTRACT

BACKGROUND: Chloride intracellular channel-1 (CLIC1) activity controls glioblastoma proliferation. Metformin exerts antitumor effects in glioblastoma stem cells (GSCs) inhibiting CLIC1 activity, but its low potency hampers its translation in clinical settings. METHODS: We synthesized a small library of novel biguanide-based compounds that were tested as antiproliferative agents for GSCs derived from human glioblastomas, in vitro using 2D and 3D cultures and in vivo in the zebrafish model. Compounds were compared to metformin for both potency and efficacy in the inhibition of GSC proliferation in vitro (MTT, Trypan blue exclusion assays, and EdU labeling) and in vivo (zebrafish model), migration (Boyden chamber assay), invasiveness (Matrigel invasion assay), self-renewal (spherogenesis assay), and CLIC1 activity (electrophysiology recordings), as well as for the absence of off-target toxicity (effects on normal stem cells and toxicity for zebrafish and chick embryos). RESULTS: We identified Q48 and Q54 as two novel CLIC1 blockers, characterized by higher antiproliferative potency than metformin in vitro, in both GSC 2D cultures and 3D spheroids. Q48 and Q54 also impaired GSC self-renewal, migration and invasion, and displayed low systemic in vivo toxicity. Q54 reduced in vivo proliferation of GSCs xenotransplanted in zebrafish hindbrain. Target specificity was confirmed by recombinant CLIC1 binding experiments using microscale thermophoresis approach. Finally, we characterized GSCs from GBMs spontaneously expressing low CLIC1 protein, demonstrating their ability to grow in vivo and to retain stem-like phenotype and functional features in vitro. In these GSCs, Q48 and Q54 displayed reduced potency and efficacy as antiproliferative agents as compared to high CLIC1-expressing tumors. However, in 3D cultures, metformin and Q48 (but not Q54) inhibited proliferation, which was dependent on the inhibition dihydrofolate reductase activity. CONCLUSIONS: These data highlight that, while CLIC1 is dispensable for the development of a subset of glioblastomas, it acts as a booster of proliferation in the majority of these tumors and its functional expression is required for biguanide antitumor class-effects. In particular, the biguanide-based derivatives Q48 and Q54, represent the leads to develop novel compounds endowed with better pharmacological profiles than metformin, to act as CLIC1-blockers for the treatment of CLIC1-expressing glioblastomas, in a precision medicine approach.


Subject(s)
Biguanides/therapeutic use , Chloride Channels/metabolism , Glioblastoma/genetics , Glioma/genetics , Neoplastic Stem Cells/metabolism , Biguanides/pharmacology , Cell Line, Tumor , Glioblastoma/pathology , Glioma/pathology , Humans
8.
Eur J Med Chem ; 229: 114054, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34959172

ABSTRACT

The human kinome plays a crucial role in several pathways. Its dysregulation has been linked to diverse central nervous system (CNS)-related disorders with a drastic impact on the aging population. Among them, tauopathies, such as Alzheimer's Disease (AD) and Frontotemporal Lobar degeneration (FTLD-tau), are neurodegenerative disorders pathologically defined by the presence of hyperphosphorylated tau-positive intracellular inclusions known as neurofibrillary tangles (NFTs). Compelling evidence has reported the great potential of the simultaneous modulation of multiple protein kinases (PKs) involved in abnormal tau phosphorylation through a concerted pharmacological approach to achieve a superior therapeutic effect relative to classic "one target, one drug" approaches. Here, we report on the identification and characterization of ARN25068 (4), a low nanomolar and well-balanced dual GSK-3ß and FYN inhibitor, which also shows inhibitory activity against DYRK1A, an emerging target in AD and tauopathies. Computational and X-Ray studies highlight compound 4's typical H-bonding pattern of ATP-competitive inhibitors at the binding sites of all three PKs. In a tau phosphorylation assay on Tau0N4R-TM-tGFP U2OS cell line, 4 reduces the extent of tau phosphorylation, promoting tau-stabilized microtubule bundles. In conclusion, this compound emerges as a promising prototype for further SAR explorations to develop potent and well-balanced triple GSK-3ß/FYN/DYRK1A inhibitors to tackle tau hyperphosphorylation.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Neuroprotective Agents/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Tauopathies/drug therapy , Binding Sites , Drug Evaluation, Preclinical , Humans , Microtubules/metabolism , Models, Molecular , Neurofibrillary Tangles/metabolism , Neuroprotective Agents/pharmacology , Phosphorylation , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship , tau Proteins/metabolism , Dyrk Kinases
9.
Drug Resist Updat ; 58: 100779, 2021 09.
Article in English | MEDLINE | ID: mdl-34461526

ABSTRACT

Pancreatic cancer is an aggressive malignancy with increasing incidence and poor prognosis due to its late diagnosis and intrinsic chemoresistance. Most pancreatic cancer patients present with locally advanced or metastatic disease characterized by inherent resistance to chemotherapy. These features pose a series of therapeutic challenges and new targets are urgently needed. Glycogen synthase kinase 3 beta (GSK3ß) is a conserved serine/threonine kinase, which regulates key cellular processes including cell proliferation, DNA repair, cell cycle progression, signaling and metabolic pathways. GSK3ß is implicated in non-malignant and malignant diseases including inflammation, neurodegenerative diseases, diabetes and cancer. GSK3ß recently emerged among the key factors involved in the onset and progression of pancreatic cancer, as well as in the acquisition of chemoresistance. Intensive research has been conducted on key oncogenic functions of GSK3ß and its potential as a druggable target; currently developed GSK3ß inhibitors display promising results in preclinical models of distinct tumor types, including pancreatic cancer. Here, we review the latest findings about GSK-3ß biology and its role in the development and progression of pancreatic cancer. Moreover, we discuss therapeutic agents targeting GSK3ß that could be administered as monotherapy or in combination with other drugs to surmount chemoresistance. Several studies are also defining potential gene signatures to identify patients who might benefit from GSK3ß-based therapeutic intervention. This detailed overview emphasizes the urgent need of additional molecular studies on the impact of GSK3ß inhibition as well as structural analysis of novel compounds and omics studies of predictive biomarkers.


Subject(s)
Drug Resistance, Neoplasm , Glycogen Synthase Kinase 3 beta/metabolism , Pancreatic Neoplasms , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Glycogen Synthase Kinase 3 beta/genetics , Humans , Oncogenes , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics
10.
Expert Opin Investig Drugs ; 28(4): 311-322, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30879349

ABSTRACT

INTRODUCTION: RX-3117 is an oral, small molecule cytidine analog anticancer agent with an improved pharmacological profile relative to gemcitabine and other nucleoside analogs. The agent has excellent activity against various cancer cell lines and xenografts including gemcitabine-resistant variants and it has excellent oral bioavailability; it is not a substrate for the degradation enzyme cytidine deaminase. RX-3117 is being evaluated at a daily oral schedule of 700 mg (5 days/week for 3 weeks) which results in plasma levels in the micromolar range that have been shown to be cytotoxic to cancer cells. It has shown clinical activity in refractory bladder cancer and pancreatic cancer. Areas covered: The review provides an overview of the relevant market and describes the mechanism of action, main pharmacokinetic/pharmacodynamic features and clinical development of this investigational small molecule. Expert opinion: RX-3117 is selectively activated by uridine-cytidine kinase 2 (UCK2), which is expressed only in tumors and has a dual mechanism of action: DNA damage and inhibition of DNA methyltransferase 1 (DNMT1). Because of its tumor selective activation, novel mechanism of action, excellent oral bioavailability and candidate biomarkers for patient selection, RX-3117 has the potential to replace gemcitabine in the treatment of a spectrum of cancer types.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Cytidine/analogs & derivatives , Neoplasms/drug therapy , Administration, Oral , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/pharmacology , Biological Availability , Cytidine/pharmacokinetics , Cytidine/pharmacology , Cytidine/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Humans , Neoplasms/pathology , Patient Selection , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...