Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Immunity ; 57(9): 2077-2094.e12, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38906145

RESUMEN

Tissues are exposed to diverse inflammatory challenges that shape future inflammatory responses. While cellular metabolism regulates immune function, how metabolism programs and stabilizes immune states within tissues and tunes susceptibility to inflammation is poorly understood. Here, we describe an innate immune metabolic switch that programs long-term intestinal tolerance. Intestinal interleukin-18 (IL-18) stimulation elicited tolerogenic macrophages by preventing their proinflammatory glycolytic polarization via metabolic reprogramming to fatty acid oxidation (FAO). FAO reprogramming was triggered by IL-18 activation of SLC12A3 (NCC), leading to sodium influx, release of mitochondrial DNA, and activation of stimulator of interferon genes (STING). FAO was maintained in macrophages by a bistable switch that encoded memory of IL-18 stimulation and by intercellular positive feedback that sustained the production of macrophage-derived 2'3'-cyclic GMP-AMP (cGAMP) and epithelial-derived IL-18. Thus, a tissue-reinforced metabolic switch encodes durable immune tolerance in the gut and may enable reconstructing compromised immune tolerance in chronic inflammation.


Asunto(s)
Tolerancia Inmunológica , Interleucina-18 , Macrófagos , Nucleótidos Cíclicos , Interleucina-18/metabolismo , Interleucina-18/inmunología , Animales , Ratones , Nucleótidos Cíclicos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Humanos , Ratones Endogámicos C57BL , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ratones Noqueados , Ácidos Grasos/metabolismo , Intestinos/inmunología , Inmunidad Innata , Inflamación/inmunología , Inflamación/metabolismo , Glucólisis , Oxidación-Reducción
2.
Chem Rev ; 123(10): 6612-6667, 2023 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-37071737

RESUMEN

The gold drugs, gold sodium thiomalate (Myocrisin), aurothioglucose (Solganal), and the orally administered auranofin (Ridaura), are utilized in modern medicine for the treatment of inflammatory arthritis including rheumatoid and juvenile arthritis; however, new gold agents have been slow to enter the clinic. Repurposing of auranofin in different disease indications such as cancer, parasitic, and microbial infections in the clinic has provided impetus for the development of new gold complexes for biomedical applications based on unique mechanistic insights differentiated from auranofin. Various chemical methods for the preparation of physiologically stable gold complexes and associated mechanisms have been explored in biomedicine such as therapeutics or chemical probes. In this Review, we discuss the chemistry of next generation gold drugs, which encompasses oxidation states, geometry, ligands, coordination, and organometallic compounds for infectious diseases, cancer, inflammation, and as tools for chemical biology via gold-protein interactions. We will focus on the development of gold agents in biomedicine within the past decade. The Review provides readers with an accessible overview of the utility, development, and mechanism of action of gold-based small molecules to establish context and basis for the thriving resurgence of gold in medicine.


Asunto(s)
Artritis Reumatoide , Auranofina , Humanos , Auranofina/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Antiinflamatorios/farmacología , Oro , Aurotioglucosa/farmacología , Aurotioglucosa/uso terapéutico , Tiomalato Sódico de Oro/farmacología , Tiomalato Sódico de Oro/uso terapéutico
3.
Angew Chem Int Ed Engl ; 63(6): e202317940, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38063406

RESUMEN

The cytotoxic and immunogenic-activating properties of a cobalt(III)-cyclam complex bearing the non-steroidal anti-inflammatory drug, flufenamic acid is reported within the context of anti-cancer stem cell (CSC) drug discovery. The cobalt(III)-cyclam complex 1 displays sub-micromolar potency towards breast CSCs grown in monolayers, 24-fold and 31-fold greater than salinomycin (an established anti-breast CSC agent) and cisplatin (an anticancer metallopharmaceutical), respectively. Strikingly, the cobalt(III)-cyclam complex 1 is 69-fold and 50-fold more potent than salinomycin and cisplatin towards three-dimensionally cultured breast CSC mammospheres. Mechanistic studies reveal that 1 induces DNA damage, inhibits cyclooxygenase-2 expression, and prompts caspase-dependent apoptosis. Breast CSCs treated with 1 exhibit damage-associated molecular patterns characteristic of immunogenic cell death and are phagocytosed by macrophages. As far as we are aware, 1 is the first cobalt complex of any oxidation state or geometry to display both cytotoxic and immunogenic-activating effects on breast CSCs.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Complejos de Coordinación , Compuestos Heterocíclicos , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Cisplatino/farmacología , Ácido Flufenámico/metabolismo , Ácido Flufenámico/farmacología , Ácido Flufenámico/uso terapéutico , Complejos de Coordinación/metabolismo , Cobalto/farmacología , Cobalto/metabolismo , Línea Celular Tumoral , Antineoplásicos/uso terapéutico , Células Madre Neoplásicas
4.
Inorg Chem ; 62(32): 13118-13129, 2023 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-37530672

RESUMEN

Diamine ligands are effective structural scaffolds for tuning the reactivity of transition-metal complexes for catalytic, materials, and phosphorescent applications and have been leveraged for biological use. In this work, we report the synthesis and characterization of a novel class of cyclometalated [C^N] Au(III) complexes bearing secondary diamines including a norbornane backbone, (2R,3S)-N2,N3-dibenzylbicyclo[2.2.1]heptane-2,3-diamine, or a cyclohexane backbone, (1R,2R)-N1,N2-dibenzylcyclohexane-1,2-diamine. X-ray crystallography confirms the square-planar geometry and chirality at nitrogen. The electronic character of the conformationally restricted norbornane backbone influences the electrochemical behavior with redox potentials of -0.8 to -1.1 V, atypical for Au(III) complexes. These compounds demonstrate promising anticancer activity, particularly, complex 1, which bears a benzylpyridine organogold framework, and supported by the bicyclic conformationally restricted diaminonorbornane, shows good potency in A2780 cells. We further show that a cellular response to 1 evokes reactive oxygen species (ROS) production and does not induce mitochondrial dysfunction. This class of complexes provides significant stability and reactivity for different applications in protein modification, catalysis, and therapeutics.


Asunto(s)
Antineoplásicos , Neoplasias Ováricas , Femenino , Humanos , Oro/farmacología , Oro/química , Antineoplásicos/química , Línea Celular Tumoral , Cristalografía por Rayos X , Diaminas/química , Norbornanos , Ligandos
5.
Chemistry ; 27(12): 4168-4175, 2021 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-33275307

RESUMEN

Direct targeting of intrinsically disordered proteins, including MYC, by small molecules for biomedical applications would resolve a longstanding issue in chemical biology and medicine. Thus, we developed gold-based small-molecule MYC reagents that engage MYC inside cells and modulate MYC transcriptional activity. Lead compounds comprise an affinity ligand and a gold(I) or gold(III) warhead capable of protein chemical modification. Cell-based MYC target engagement studies via CETSA and co-immunoprecipitation reveal specific interaction of compounds with MYC in cells. The lead gold(I) reagent, 1, demonstrates superior cell-killing potential (up to 35-fold) in a MYC-dependent manner when compared to 10058-F4 in cells including the TNBC, MDA-MB-231. Subsequently, 1 suppresses MYC transcription factor activity via functional colorimetric assays, and gene-profiling using whole-cell transcriptomics reveals significant modulation of MYC target genes by 1. These findings point to metal-mediated ligand affinity chemistry (MLAC) based on gold as a promising strategy to develop chemical probes and anticancer therapeutics targeting MYC.


Asunto(s)
Oro , Proteínas Proto-Oncogénicas c-myc , Apoptosis , Línea Celular Tumoral , Regulación de la Expresión Génica , Ligandos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo
6.
Inorg Chem ; 60(19): 14582-14593, 2021 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-34402302

RESUMEN

Transition-metal-based approaches to selectively modify proteins hold promise in addressing challenges in chemical biology. Unique bioorthogonal chemistry can be achieved with preformed metal-based compounds; however, their utility in native protein sites within cells remain underdeveloped. Here, we tune the ancillary ligands of cyclometalated gold(III) as a reactive group, and the gold scaffold allows for rapid modification of a desired cysteine residue proximal to the ligand binding site of a target protein. Moreover, evidence for a ligand association mechanism toward C-S bond formation by X-crystallography is established. The observed reactivity of cyclometalated gold(III) enables the rational design of a cysteine-targeted covalent inhibitor of mutant KRAS. This work illustrates the potential of structure-activity relationship studies to tune kinetics of cysteine arylation and rational design of metal-mediated ligand affinity chemistry (MLAC) of native proteins.


Asunto(s)
Cisteína/farmacología , Inhibidores Enzimáticos/farmacología , Oro/farmacología , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Cisteína/química , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Oro/química , Humanos , Ligandos , Estructura Molecular , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(5): 950-955, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28096358

RESUMEN

Cisplatin is the most commonly used anticancer drug for the treatment of testicular germ cell tumors (TGCTs). The hypersensitivity of TGCTs to cisplatin is a subject of widespread interest. Here, we show that high-mobility group box protein 4 (HMGB4), a protein preferentially expressed in testes, uniquely blocks excision repair of cisplatin-DNA adducts, 1,2-intrastrand cross-links, to potentiate the sensitivity of TGCTs to cisplatin therapy. We used CRISPR/Cas9-mediated gene editing to knockout the HMGB4 gene in a testicular human embryonic carcinoma and examined cellular responses. We find that loss of HMGB4 elicits resistance to cisplatin as evidenced by cell proliferation and apoptosis assays. We demonstrate that HMGB4 specifically inhibits repair of the major cisplatin-DNA adducts in TGCT cells by using the human TGCT excision repair system. Our findings also reveal characteristic HMGB4-dependent differences in cell cycle progression following cisplatin treatment. Collectively, these data provide convincing evidence that HMGB4 plays a major role in sensitizing TGCTs to cisplatin, consistent with shielding of platinum-DNA adducts from excision repair.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Reparación del ADN , Proteínas HMGB/metabolismo , Neoplasias de Células Germinales y Embrionarias/metabolismo , Neoplasias Testiculares/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Daño del ADN , Edición Génica , Proteínas HMGB/genética , Humanos
8.
Inorganica Chim Acta ; 503(1)2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34565828

RESUMEN

Cancer remains one of the leading causes of death worldwide and despite several attempts using chemotherapy to combat the deadly disease, toxic side effects and drug resistance temper efficacy [1]. Thus, drugs with potentially new mechanisms and lower toxicity to normal cells are needed. Metalloids such as arsenic compounds have been clinically beneficial in fighting cancer, but germanium is yet to gain such prominence [2,3]. We report the synthesis of four octahedral germanium(IV) complexes bearing acetylacetonato ligand, [GeIV(acac)3)]+, with different anions (3 - 6) using a streamlined synthetic approach. The compounds were structurally and electrochemically characterized using NMR, MS, X-ray crystallography, and cyclic voltammetry. The cyclic voltammogram of 3-5 revealed distinct irreversible peaks in the range of -0.9 to -1.9 V, corresponding to Ge(IV)/ Ge(II) or Ge(II)/Ge(0) couple in DMSO. We explored the anticancer activity of the complexes against a panel of cancer cell lines with IC50 values in the sub-micromolar range (9-15 µM). The compounds display ~3-fold selectivity in cancer cells over normal epithelial cells. In addition to the promising anticancer activity, the compounds display high complex stability in biological media, induces G1 arrest, reactive oxygen stress (ROS) accumulation, and mitochondria membrane depolarization in cancer cells. Furthermore, the compounds induce significant apoptosis.

9.
Molecules ; 25(23)2020 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-33291802

RESUMEN

Herein is reported the synthesis of two Au(III) complexes bearing the (R,R)-(-)-2,3-Bis(tert-butylmethylphosphino)quinoxaline (R,R-QuinoxP*) or (S,S)-(+)-2,3-Bis(tert-butylmethylphosphino)quinoxaline (S,S-QuinoxP*) ligands. By reacting two stoichiometric equivalents of HAuCl4.3H2O to one equivalent of the corresponding QuinoxP* ligand, (R,R)-(-)-2,3-Bis(tert-butylmethylphosphino)quinoxalinedichlorogold(III) tetrachloroaurates(III) (1) and (S,S)-(+)-2,3-Bis(tert-butylmethylphosphino)quinoxalinedichlorogold(III) tetrachloroaurates(III) (2) were formed, respectively, in moderate yields. The structure of (S,S)-(+)-2,3-Bis(tert-butylmethylphosphino)quinoxalinedichlorogold(III) tetrachloroaurates(III) (2) was further confirmed by X-ray crystallography. The antiproliferative activities of the two compounds were evaluated in a panel of cell lines and exhibited promising results comparable to auranofin and cisplatin with IC50 values between 1.08 and 4.83 µM. It is noteworthy that in comparison to other platinum and ruthenium enantiomeric complexes, the two enantiomers (1 and 2) do not exhibit different cytotoxic effects. The compounds exhibited stability in biologically relevant media over 48 h as well as inert reactivity to excess glutathione at 37 °C. These results demonstrate that the Au(III) atom, stabilized by the QuinoxP* ligand, can provide exciting compounds for novel anticancer drugs. These complexes provide a new scaffold to further develop a robust and diverse library of chiral phosphorus Au(III) complexes.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Oro/química , Oro/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X/métodos , Glutatión/química , Humanos , Ligandos , Estereoisomerismo
10.
Inorg Chem ; 58(14): 9326-9340, 2019 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-31247820

RESUMEN

The synthesis of a novel class of cyclometalated gold(III) complexes supported by benzoylpyridine, benzylpyridine, and (1R,2R)-(+)-1,2-diaminocyclohexane (DACH) ligands, along with their crystal structures, is reported. These compounds provide a new scaffold to investigate biological properties of gold(III) complexes. The six complexes were prepared and characterized, following reactions of (C,N) cyclometalated gold(III) scaffolds, [Au(C^N)Cl2] with DACH, which yielded a new series of cyclometaled gold(III), 3-5, of the type [Au(C^NH)(DACH)2]+ and the nitrogen-substituted cyclometalated Au(III), 6-8, of the type [Au(C^N)(DACH)]2+. Antiproliferative activity of these complexes in a panel of cancer cells showed promising results with IC50 in the micromolar range and selectivity over normal epithelial cells, MRC5. Whereas 8 shows minimal interaction with superhelical DNA except at high gold concentrations of 500 µM, complex 5 does not show interaction even at 1000 µM. The complexes display significant uptake in OVCAR8 cancer cells within 200-1200 pmol/million cells with the exception of complex 4. Differential cellular uptake was observed for the complexes; for example, while 3 and 8 display significant uptake, 4 showed minimal uptake. The compounds proved to be stable under physiological conditions and were minimally affected by either glutathione or sodium ascorbate. Cell cycle studies reveal a G1 arrest induced by representative complexes. The results reveal that enhanced Au(III) stabilization promoted by combined cyclometalated and DACH ligands may offer ligand tuning insights for novel anticancer drug design.


Asunto(s)
Ciclohexilaminas/química , Ciclohexilaminas/farmacología , Compuestos de Oro/química , Compuestos de Oro/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Ácido Ascórbico/química , Línea Celular Tumoral , Simulación por Computador , Cristalografía por Rayos X , Glutatión/química , Humanos , Ligandos , Modelos Químicos , Modelos Moleculares , Estructura Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA