Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Nat Commun ; 15(1): 3827, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38714735

RESUMO

The main protease (Mpro) of SARS-CoV-2 is critical for viral function and a key drug target. Mpro is only active when reduced; turnover ceases upon oxidation but is restored by re-reduction. This suggests the system has evolved to survive periods in an oxidative environment, but the mechanism of this protection has not been confirmed. Here, we report a crystal structure of oxidized Mpro showing a disulfide bond between the active site cysteine, C145, and a distal cysteine, C117. Previous work proposed this disulfide provides the mechanism of protection from irreversible oxidation. Mpro forms an obligate homodimer, and the C117-C145 structure shows disruption of interactions bridging the dimer interface, implying a correlation between oxidation and dimerization. We confirm dimer stability is weakened in solution upon oxidation. Finally, we observe the protein's crystallization behavior is linked to its redox state. Oxidized Mpro spontaneously forms a distinct, more loosely packed lattice. Seeding with crystals of this lattice yields a structure with an oxidation pattern incorporating one cysteine-lysine-cysteine (SONOS) and two lysine-cysteine (NOS) bridges. These structures further our understanding of the oxidative regulation of Mpro and the crystallization conditions necessary to study this structurally.


Assuntos
Domínio Catalítico , Proteases 3C de Coronavírus , Cisteína , Dissulfetos , Oxirredução , SARS-CoV-2 , Dissulfetos/química , Dissulfetos/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/química , Proteases 3C de Coronavírus/metabolismo , Proteases 3C de Coronavírus/química , Cisteína/química , Cisteína/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Multimerização Proteica , COVID-19/virologia
2.
J Mol Biol ; 436(5): 168439, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38185322

RESUMO

The understanding of signal transduction mechanisms in photoreceptor proteins is essential for elucidating how living organisms respond to light as environmental stimuli. In this study, we investigated the ATP binding, photoactivation and signal transduction process in the photoactivatable adenylate cyclase from Oscillatoria acuminata (OaPAC) upon blue light excitation. Structural models with ATP bound in the active site of native OaPAC at cryogenic as well as room temperature are presented. ATP is found in one conformation at cryogenic- and in two conformations at ambient-temperature, and is bound in an energetically unfavorable conformation for the conversion to cAMP. However, FTIR spectroscopic experiments confirm that this conformation is the native binding mode in dark state OaPAC and that transition to a productive conformation for ATP turnover only occurs after light activation. A combination of time-resolved crystallography experiments at synchrotron and X-ray Free Electron Lasers sheds light on the early events around the Flavin Adenine Dinucleotide (FAD) chromophore in the light-sensitive BLUF domain of OaPAC. Early changes involve the highly conserved amino acids Tyr6, Gln48 and Met92. Crucially, the Gln48 side chain performs a 180° rotation during activation, leading to the stabilization of the FAD chromophore. Cryo-trapping experiments allowed us to investigate a late light-activated state of the reaction and revealed significant conformational changes in the BLUF domain around the FAD chromophore. In particular, a Trpin/Metout transition upon illumination is observed for the first time in the BLUF domain and its role in signal transmission via α-helix 3 and 4 in the linker region between sensor and effector domain is discussed.


Assuntos
Adenilil Ciclases , Proteínas de Bactérias , Oscillatoria , Fotorreceptores Microbianos , Trifosfato de Adenosina/química , Adenilil Ciclases/química , Adenilil Ciclases/efeitos da radiação , Proteínas de Bactérias/química , Proteínas de Bactérias/efeitos da radiação , Flavina-Adenina Dinucleotídeo/química , Transdução de Sinais , Espectroscopia de Infravermelho com Transformada de Fourier , Oscillatoria/enzimologia , Domínio Catalítico , Triptofano/química , Metionina/química , Fotorreceptores Microbianos/química , Fotorreceptores Microbianos/efeitos da radiação , Ativação Enzimática
3.
Nature ; 625(7994): 385-392, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38123683

RESUMO

Digested dietary fats are taken up by enterocytes where they are assembled into pre-chylomicrons in the endoplasmic reticulum followed by transport to the Golgi for maturation and subsequent secretion to the circulation1. The role of mitochondria in dietary lipid processing is unclear. Here we show that mitochondrial dysfunction in enterocytes inhibits chylomicron production and the transport of dietary lipids to peripheral organs. Mice with specific ablation of the mitochondrial aspartyl-tRNA synthetase DARS2 (ref. 2), the respiratory chain subunit SDHA3 or the assembly factor COX10 (ref. 4) in intestinal epithelial cells showed accumulation of large lipid droplets (LDs) in enterocytes of the proximal small intestine and failed to thrive. Feeding a fat-free diet suppressed the build-up of LDs in DARS2-deficient enterocytes, which shows that the accumulating lipids derive mostly from digested fat. Furthermore, metabolic tracing studies revealed an impaired transport of dietary lipids to peripheral organs in mice lacking DARS2 in intestinal epithelial cells. DARS2 deficiency caused a distinct lack of mature chylomicrons concomitant with a progressive dispersal of the Golgi apparatus in proximal enterocytes. This finding suggests that mitochondrial dysfunction results in impaired trafficking of chylomicrons from the endoplasmic reticulum to the Golgi, which in turn leads to storage of dietary lipids in large cytoplasmic LDs. Taken together, these results reveal a role for mitochondria in dietary lipid transport in enterocytes, which might be relevant for understanding the intestinal defects observed in patients with mitochondrial disorders5.


Assuntos
Gorduras na Dieta , Enterócitos , Metabolismo dos Lipídeos , Mitocôndrias , Animais , Camundongos , Aspartato-tRNA Ligase/metabolismo , Quilomícrons/metabolismo , Gorduras na Dieta/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Retículo Endoplasmático/metabolismo , Enterócitos/metabolismo , Enterócitos/patologia , Células Epiteliais/metabolismo , Complexo de Golgi/metabolismo , Intestinos , Gotículas Lipídicas/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia
4.
Eur J Med Chem ; 259: 115670, 2023 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-37515920

RESUMO

Alzheimer's disease (AD) is a progressive brain disorder associated with slow loss of brain functions leading to memory failure and modest changes in behavior. The multifactorial neuropathological condition is due to a depletion of cholinergic neurons and accumulation of amyloid-beta (Aß) plaques. Recently, a multi-target-directed ligand (MTDL) strategy has emerged as a robust drug discovery tool to overcome current challenges. In this research work, we aimed to design and develop a library of triazole-bridged aryl adamantane analogs for the treatment of AD. All synthesized analogs were characterized and evaluated through various in vitro and in vivo biological studies. The optimal compounds 32 and 33 exhibited potent inhibitory activities against acetylcholinesterase (AChE) (32 - IC50 = 0.086 µM; 33 - 0.135 µM), and significant Aß aggregation inhibition (20 µM). N-methyl-d-aspartate (NMDA) receptor (GluN1-1b/GluN2B subunit combination) antagonistic activity of compounds 32 and 33 measured upon heterologous expression in Xenopus laevis oocytes showed IC50 values of 3.00 µM and 2.86 µM, respectively. The compounds possessed good blood-brain barrier permeability in the PAMPA assay and were safe for SH-SY5Y neuroblastoma (10 µM) and HEK-293 cell lines (30 µM). Furthermore, in vivo behavioral studies in rats demonstrated that both compounds improved cognitive and spatial memory impairment at a dose of 10 mg/kg oral administration. Together, our findings suggest triazole-bridged aryl adamantane as a promising new scaffold for the development of anti-Alzheimer's drugs.


Assuntos
Doença de Alzheimer , Neuroblastoma , Fármacos Neuroprotetores , Triazóis , Animais , Humanos , Ratos , Acetilcolinesterase/metabolismo , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Inibidores da Colinesterase/farmacologia , Inibidores da Colinesterase/uso terapêutico , Desenho de Fármacos , Células HEK293 , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Relação Estrutura-Atividade , Triazóis/química , Triazóis/farmacologia
5.
Leukemia ; 37(8): 1611-1625, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37414921

RESUMO

Venetoclax/azacitidine combination therapy is effective in acute myeloid leukemia (AML) and tolerable for older, multimorbid patients. Despite promising response rates, many patients do not achieve sustained remission or are upfront refractory. Identification of resistance mechanisms and additional therapeutic targets represent unmet clinical needs. By using a genome-wide CRISPR/Cas9 library screen targeting 18,053 protein- coding genes in a human AML cell line, various genes conferring resistance to combined venetoclax/azacitidine treatment were identified. The ribosomal protein S6 kinase A1 (RPS6KA1) was among the most significantly depleted sgRNA-genes in venetoclax/azacitidine- treated AML cells. Addition of the RPS6KA1 inhibitor BI-D1870 to venetoclax/azacitidine decreased proliferation and colony forming potential compared to venetoclax/azacitidine alone. Furthermore, BI-D1870 was able to completely restore the sensitivity of OCI-AML2 cells with acquired resistance to venetoclax/azacitidine. Analysis of cell surface markers revealed that RPS6KA1 inhibition efficiently targeted monocytic blast subclones as a potential source of relapse upon venetoclax/azacitidine treatment. Taken together, our results suggest RPS6KA1 as mediator of resistance towards venetoclax/azacitidine and additional RPS6KA1 inhibition as strategy to prevent or overcome resistance.


Assuntos
Azacitidina , Resistencia a Medicamentos Antineoplásicos , Leucemia Mieloide Aguda , Humanos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Azacitidina/uso terapêutico , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Proteínas Quinases S6 Ribossômicas , Proteínas Quinases S6 Ribossômicas 90-kDa , RNA Guia de Sistemas CRISPR-Cas
6.
Cell Rep ; 42(7): 112751, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37405921

RESUMO

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a cancer syndrome caused by inactivating germline mutations in fumarate hydratase (FH) and subsequent accumulation of fumarate. Fumarate accumulation leads to profound epigenetic changes and the activation of an anti-oxidant response via nuclear translocation of the transcription factor NRF2. The extent to which chromatin remodeling shapes this anti-oxidant response is currently unknown. Here, we explored the effects of FH loss on the chromatin landscape to identify transcription factor networks involved in the remodeled chromatin landscape of FH-deficient cells. We identify FOXA2 as a key transcription factor that regulates anti-oxidant response genes and subsequent metabolic rewiring cooperating without direct interaction with the anti-oxidant regulator NRF2. The identification of FOXA2 as an anti-oxidant regulator provides additional insights into the molecular mechanisms behind cell responses to fumarate accumulation and potentially provides further avenues for therapeutic intervention for HLRCC.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Leiomiomatose , Síndromes Neoplásicas Hereditárias , Neoplasias Cutâneas , Neoplasias Uterinas , Feminino , Humanos , Fumarato Hidratase/genética , Antioxidantes , Fator 2 Relacionado a NF-E2/genética , Leiomiomatose/genética , Neoplasias Uterinas/genética , Neoplasias Cutâneas/genética , Síndromes Neoplásicas Hereditárias/genética , Cromatina , Neoplasias Renais/genética , Carcinoma de Células Renais/genética , Fator 3-beta Nuclear de Hepatócito/genética
7.
Nat Cardiovasc Res ; 2(8): 733-745, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38666037

RESUMO

Recurrent myocardial ischemia can lead to left ventricular (LV) dysfunction in patients with coronary artery disease (CAD). In this observational cohort study, we assessed for chronic metabolomic and transcriptomic adaptations within LV myocardium of patients undergoing coronary artery bypass grafting. During surgery, paired transmural LV biopsies were acquired on the beating heart from regions with and without evidence of inducible ischemia on preoperative stress perfusion cardiovascular magnetic resonance. From 33 patients, 63 biopsies were acquired, compared to analysis of LV samples from 11 donor hearts. The global myocardial adenosine triphosphate (ATP):adenosine diphosphate (ADP) ratio was reduced in patients with CAD as compared to donor LV tissue, with increased expression of oxidative phosphorylation (OXPHOS) genes encoding the electron transport chain complexes across multiple cell types. Paired analyses of biopsies obtained from LV segments with or without inducible ischemia revealed no significant difference in the ATP:ADP ratio, broader metabolic profile or expression of ventricular cardiomyocyte genes implicated in OXPHOS. Differential metabolite analysis suggested dysregulation of several intermediates in patients with reduced LV ejection fraction, including succinate. Overall, our results suggest that viable myocardium in patients with stable CAD has global alterations in bioenergetic and transcriptional profile without large regional differences between areas with or without inducible ischemia.

8.
Nat Commun ; 13(1): 7830, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36539415

RESUMO

Metabolic reprogramming is critical for tumor initiation and progression. However, the exact impact of specific metabolic changes on cancer progression is poorly understood. Here, we integrate multimodal analyses of primary and metastatic clonally-related clear cell renal cancer cells (ccRCC) grown in physiological media to identify key stage-specific metabolic vulnerabilities. We show that a VHL loss-dependent reprogramming of branched-chain amino acid catabolism sustains the de novo biosynthesis of aspartate and arginine enabling tumor cells with the flexibility of partitioning the nitrogen of the amino acids depending on their needs. Importantly, we identify the epigenetic reactivation of argininosuccinate synthase (ASS1), a urea cycle enzyme suppressed in primary ccRCC, as a crucial event for metastatic renal cancer cells to acquire the capability to generate arginine, invade in vitro and metastasize in vivo. Overall, our study uncovers a mechanism of metabolic flexibility occurring during ccRCC progression, paving the way for the development of novel stage-specific therapies.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Humanos , Carcinoma de Células Renais/genética , Aminoácidos de Cadeia Ramificada , Nitrogênio , Neoplasias Renais/genética , Arginina/metabolismo , Linhagem Celular Tumoral
9.
Obstet Gynecol ; 140(5): 861-868, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36201759

RESUMO

OBJECTIVE: To characterize current trends and outcomes in pregnancies complicated by cystic fibrosis (CF) that resulted in delivery. METHODS: This repeated cross-sectional study used the U.S. National Inpatient Sample to identify delivery hospitalizations of patients with CF between 2000 and 2019. Trends in delivery hospitalizations of patients with CF were assessed using joinpoint regression to determine the average annual percent change (AAPC). The risk of adverse maternal and obstetric outcomes was compared between patients with and without CF using adjusted logistic regression models accounting for demographic, clinical, and hospital characteristics, with adjusted odds ratios (aORs) with 95% CIs as measures of association. The proportion of patients with CF and other chronic conditions such as pregestational diabetes was analyzed over time. RESULTS: From 2000 to 2019, the prevalence of CF at delivery increased from 2.1 to 10.4 per 100,000 deliveries (AAPC 6.7%, 95% CI 5.7-8.2%). The proportion of patients with CF and other chronic conditions increased from 18.0% to 37.3% (AAPC 3.1%, 95% CI 1.0-5.3%). Patients with CF were more likely to experience severe maternal morbidity (aOR 2.61, 95% CI 1.71-3.97), respiratory complications (aOR 17.45, 95% CI 11.85-25.68), venous thromboembolism (aOR 3.59, 95% CI 1.33-9.69), preterm delivery (aOR 2.15, 95% CI 1.79-2.59), abruption and antepartum hemorrhage (aOR 1.63, 95% CI 1.10-2.41), and gestational diabetes (aOR 2.47, 95% CI 2.47-3.70). CONCLUSION: Although still infrequent (approximately 1 in 10,000), deliveries complicated by CF increased approximately fivefold over the study period. The proportion of patients with CF and other chronic conditions is increasing. Patients with CF are at increased risk for a broad range of adverse outcomes.


Assuntos
Fibrose Cística , Complicações do Trabalho de Parto , Complicações na Gravidez , Nascimento Prematuro , Humanos , Gravidez , Recém-Nascido , Feminino , Complicações na Gravidez/epidemiologia , Fibrose Cística/complicações , Fibrose Cística/epidemiologia , Estudos Transversais , Complicações do Trabalho de Parto/epidemiologia , Nascimento Prematuro/epidemiologia , Estudos Retrospectivos
10.
Sci Adv ; 8(42): eabq8297, 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36269833

RESUMO

Fumarate hydratase (FH) is a mitochondrial enzyme that catalyzes the reversible hydration of fumarate to malate in the tricarboxylic acid (TCA) cycle. Germline mutations of FH lead to hereditary leiomyomatosis and renal cell carcinoma (HLRCC), a cancer syndrome characterized by a highly aggressive form of renal cancer. Although HLRCC tumors metastasize rapidly, FH-deficient mice develop premalignant cysts in the kidneys, rather than carcinomas. How Fh1-deficient cells overcome these tumor-suppressive events during transformation is unknown. Here, we perform a genome-wide CRISPR-Cas9 screen to identify genes that, when ablated, enhance the proliferation of Fh1-deficient cells. We found that the depletion of the histone cell cycle regulator (HIRA) enhances proliferation and invasion of Fh1-deficient cells in vitro and in vivo. Mechanistically, Hira loss activates MYC and its target genes, increasing nucleotide metabolism specifically in Fh1-deficient cells, independent of its histone chaperone activity. These results are instrumental for understanding mechanisms of tumorigenesis in HLRCC and the development of targeted treatments for patients.

11.
Commun Biol ; 5(1): 805, 2022 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-35953531

RESUMO

SARS-CoV-2 papain-like protease (PLpro) covers multiple functions. Beside the cysteine-protease activity, facilitating cleavage of the viral polypeptide chain, PLpro has the additional and vital function of removing ubiquitin and ISG15 (Interferon-stimulated gene 15) from host-cell proteins to support coronaviruses in evading the host's innate immune responses. We identified three phenolic compounds bound to PLpro, preventing essential molecular interactions to ISG15 by screening a natural compound library. The compounds identified by X-ray screening and complexed to PLpro demonstrate clear inhibition of PLpro in a deISGylation activity assay. Two compounds exhibit distinct antiviral activity in Vero cell line assays and one inhibited a cytopathic effect in non-cytotoxic concentration ranges. In the context of increasing PLpro mutations in the evolving new variants of SARS-CoV-2, the natural compounds we identified may also reinstate the antiviral immune response processes of the host that are down-regulated in COVID-19 infections.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , Sítio Alostérico , Antivirais/farmacologia , Proteases Semelhantes à Papaína de Coronavírus , Humanos , Papaína/metabolismo , Peptídeo Hidrolases/metabolismo , SARS-CoV-2
12.
Blood ; 140(24): 2594-2610, 2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-35857899

RESUMO

BCL-2 inhibition has been shown to be effective in acute myeloid leukemia (AML) in combination with hypomethylating agents or low-dose cytarabine. However, resistance and relapse represent major clinical challenges. Therefore, there is an unmet need to overcome resistance to current venetoclax-based strategies. We performed high-throughput drug screening to identify effective combination partners for venetoclax in AML. Overall, 64 antileukemic drugs were screened in 31 primary high-risk AML samples with or without venetoclax. Gilteritinib exhibited the highest synergy with venetoclax in FLT3 wild-type AML. The combination of gilteritinib and venetoclax increased apoptosis, reduced viability, and was active in venetoclax-azacitidine-resistant cell lines and primary patient samples. Proteomics revealed increased FLT3 wild-type signaling in specimens with low in vitro response to the currently used venetoclax-azacitidine combination. Mechanistically, venetoclax with gilteritinib decreased phosphorylation of ERK and GSK3B via combined AXL and FLT3 inhibition with subsequent suppression of the antiapoptotic protein MCL-1. MCL-1 downregulation was associated with increased MCL-1 phosphorylation of serine 159, decreased phosphorylation of threonine 161, and proteasomal degradation. Gilteritinib and venetoclax were active in an FLT3 wild-type AML patient-derived xenograft model with TP53 mutation and reduced leukemic burden in 4 patients with FLT3 wild-type AML receiving venetoclax-gilteritinib off label after developing refractory disease under venetoclax-azacitidine. In summary, our results suggest that combined inhibition of FLT3/AXL potentiates venetoclax response in FLT3 wild-type AML by inducing MCL-1 degradation. Therefore, the venetoclax-gilteritinib combination merits testing as a potentially active regimen in patients with high-risk FLT3 wild-type AML.


Assuntos
Leucemia Mieloide Aguda , Humanos , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Azacitidina , Tirosina Quinase 3 Semelhante a fms/genética
13.
J Cell Biol ; 221(7)2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35670764

RESUMO

Fbxo7 is associated with cancer and Parkinson's disease. Although Fbxo7 recruits substrates for SCF-type ubiquitin ligases, it also promotes Cdk6 activation in a ligase-independent fashion. We discovered PFKP, the gatekeeper of glycolysis, in a screen for Fbxo7 substrates. PFKP is an essential Cdk6 substrate in some T-ALL cells. We investigated the molecular relationship between Fbxo7, Cdk6, and PFKP, and the effect of Fbxo7 on T cell metabolism, viability, and activation. Fbxo7 promotes Cdk6-independent ubiquitination and Cdk6-dependent phosphorylation of PFKP. Importantly, Fbxo7-deficient cells have reduced Cdk6 activity, and hematopoietic and lymphocytic cells show high expression and significant dependency on Fbxo7. CD4+ T cells with reduced Fbxo7 show increased glycolysis, despite lower cell viability and activation levels. Metabolomic studies of activated CD4+ T cells confirm increased glycolytic flux in Fbxo7-deficient cells, alongside altered nucleotide biosynthesis and arginine metabolism. We show Fbxo7 expression is glucose-responsive at the mRNA and protein level and propose Fbxo7 inhibits PFKP and glycolysis via its activation of Cdk6.


Assuntos
Quinase 6 Dependente de Ciclina , Proteínas F-Box , Fosfofrutoquinase-1 Tipo C , Linfócitos T , Quinase 6 Dependente de Ciclina/metabolismo , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Glicólise , Humanos , Fosfofrutoquinase-1 Tipo C/metabolismo , Linfócitos T/metabolismo , Ubiquitinação
14.
EMBO Mol Med ; 14(4): e14990, 2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-35253392

RESUMO

The heterogeneous response of acute myeloid leukemia (AML) to current anti-leukemic therapies is only partially explained by mutational heterogeneity. We previously identified GPR56 as a surface marker associated with poor outcome across genetic groups, which characterizes two leukemia stem cell (LSC)-enriched compartments with different self-renewal capacities. How these compartments self-renew remained unclear. Here, we show that GPR56+ LSC compartments are promoted in a complex network involving epithelial-to-mesenchymal transition (EMT) regulators besides Rho, Wnt, and Hedgehog (Hh) signaling. Unexpectedly, Wnt pathway inhibition increased the more immature, slowly cycling GPR56+ CD34+ fraction and Hh/EMT gene expression, while Wnt activation caused opposite effects. Our data suggest that the crucial role of GPR56 lies in its ability to co-activate these opposing signals, thus ensuring the constant supply of both LSC subsets. We show that CDK7 inhibitors suppress both LSC-enriched subsets in vivo and synergize with the Bcl-2 inhibitor venetoclax. Our data establish reciprocal transition between LSC compartments as a novel concept underlying the poor outcome in GPR56high AML and propose combined CDK7 and Bcl-2 inhibition as LSC-directed therapy in this disease.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes , Quinases Ciclina-Dependentes , Leucemia Mieloide Aguda , Inibidores de Proteínas Quinases , Sulfonamidas , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Proteína Quinase CDC2/antagonistas & inibidores , Quinases Ciclina-Dependentes/antagonistas & inibidores , Sinergismo Farmacológico , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/uso terapêutico , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/uso terapêutico , Sulfonamidas/farmacologia , Quinase Ativadora de Quinase Dependente de Ciclina
15.
Cells ; 11(4)2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35203403

RESUMO

In a prospective observational pilot study on patients undergoing elective cardiac surgery with cardiopulmonary bypass, we evaluated label-free quantitative phase imaging (QPI) with digital holographic microscopy (DHM) to describe perioperative inflammation by changes in biophysical cell properties of lymphocytes and monocytes. Blood samples from 25 patients were investigated prior to cardiac surgery and postoperatively at day 1, 3 and 6. Biophysical and morphological cell parameters accessible with DHM, such as cell volume, refractive index, dry mass, and cell shape related form factor, were acquired and compared to common flow cytometric blood cell markers of inflammation and selected routine laboratory parameters. In all examined patients, cardiac surgery induced an acute inflammatory response as indicated by changes in routine laboratory parameters and flow cytometric cell markers. DHM results were associated with routine laboratory and flow cytometric data and correlated with complications in the postoperative course. In a subgroup analysis, patients were classified according to the inflammation related C-reactive protein (CRP) level, treatment with epinephrine and the occurrence of postoperative complications. Patients with regular courses, without epinephrine treatment and with low CRP values showed a postoperative lymphocyte volume increase. In contrast, the group of patients with increased CRP levels indicated an even further enlarged lymphocyte volume, while for the groups of epinephrine treated patients and patients with complicative courses, no postoperative lymphocyte volume changes were detected. In summary, the study demonstrates the capability of DHM to describe biophysical cell parameters of perioperative lymphocytes and monocytes changes in cardiac surgery patients. The pattern of correlations between biophysical DHM data and laboratory parameters, flow cytometric cell markers, and the postoperative course exemplify DHM as a promising diagnostic tool for a characterization of inflammatory processes and course of disease.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Microscopia , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Epinefrina , Humanos , Inflamação , Microscopia/métodos , Monócitos , Estudos Prospectivos
16.
Int J Cancer ; 150(7): 1141-1155, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-34766343

RESUMO

Immunotherapies, such as chimeric antigen receptor (CAR) modified T cells and antibody-drug conjugates (ADCs), have revolutionized the treatment of cancer, especially of lymphoid malignancies. The application of targeted immunotherapy to patients with acute myeloid leukemia (AML) has been limited in particular by the lack of a tumor-specific target antigen. Gemtuzumab ozogamicin (GO), an ADC targeting CD33, is the only approved immunotherapeutic agent in AML. In our study, we introduce a CD33-directed third-generation CAR T-cell product (3G.CAR33-T) for the treatment of patients with AML. 3G.CAR33-T cells could be expanded up to the end-of-culture, that is, 17 days after transduction, and displayed significant cytokine secretion and robust cytotoxic activity when incubated with CD33-positive cells including cell lines, drug-resistant cells, primary blasts as well as normal hematopoietic stem and progenitor cells (HSPCs). When compared to second-generation CAR33-T cells, 3G.CAR33-T cells exhibited higher viability, increased proliferation and stronger cytotoxicity. Also, GO exerted strong antileukemia activity against CD33-positive AML cells. Upon genomic deletion of CD33 in HSPCs, 3G.CAR33-T cells and GO preferentially killed wildtype leukemia cells, while sparing CD33-deficient HSPCs. Our data provide evidence for the applicability of CD33-targeted immunotherapies in AML and its potential implementation in CD33 genome-edited stem cell transplantation approaches.


Assuntos
Gemtuzumab/uso terapêutico , Transplante de Células-Tronco Hematopoéticas , Imunoterapia Adotiva , Leucemia Mieloide Aguda/terapia , Receptores de Antígenos Quiméricos/imunologia , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Edição de Genes , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/patologia , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/análise , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/genética
17.
J Sex Med ; 18(12): 1945-1949, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34688584

RESUMO

There has been an increase in genital cosmentic surgeries over the past decade, with the most common procedure being labiaplasty. This trend has many origins, but a significant motivator is genital self image, which has been shown to be very culturally influenced. Furthermore genital self modification, by way of grooming also is felt to play a role in the desire to surgically alter one's genital appearance. Given the cultural aspect of these practices, sexual health proivders should be aware of the role of self image and self modification in the drive towards persuing surgical changes to the genitals. Schmidt CN, Rowen TS. Female Genital Self-Image and Modification. J Sex Med 2021;18:1945-1949.


Assuntos
Imagem Corporal , Autoimagem , Emoções , Feminino , Genitália , Genitália Feminina/cirurgia , Humanos
18.
Leukemia ; 35(7): 2002-2016, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33953289

RESUMO

B cells have the unique property to somatically alter their immunoglobulin (IG) genes by V(D)J recombination, somatic hypermutation (SHM) and class-switch recombination (CSR). Aberrant targeting of these mechanisms is implicated in lymphomagenesis, but the mutational processes are poorly understood. By performing whole genome and transcriptome sequencing of 181 germinal center derived B-cell lymphomas (gcBCL) we identified distinct mutational signatures linked to SHM and CSR. We show that not only SHM, but presumably also CSR causes off-target mutations in non-IG genes. Kataegis clusters with high mutational density mainly affected early replicating regions and were enriched for SHM- and CSR-mediated off-target mutations. Moreover, they often co-occurred in loci physically interacting in the nucleus, suggesting that mutation hotspots promote increased mutation targeting of spatially co-localized loci (termed hypermutation by proxy). Only around 1% of somatic small variants were in protein coding sequences, but in about half of the driver genes, a contribution of B-cell specific mutational processes to their mutations was found. The B-cell-specific mutational processes contribute to both lymphoma initiation and intratumoral heterogeneity. Overall, we demonstrate that mutational processes involved in the development of gcBCL are more complex than previously appreciated, and that B cell-specific mutational processes contribute via diverse mechanisms to lymphomagenesis.


Assuntos
Genoma/genética , Centro Germinativo/metabolismo , Linfoma de Células B/genética , Mutação/genética , Adulto , Linfócitos B/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Genes de Imunoglobulinas/genética , Células HeLa , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana , Humanos , Switching de Imunoglobulina/genética , Células K562 , Células MCF-7 , Hipermutação Somática de Imunoglobulina/genética , Recombinação V(D)J/genética
19.
Mol Psychiatry ; 26(6): 2471-2482, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32089545

RESUMO

Circulating autoantibodies (AB) of different immunoglobulin classes (IgM, IgA, and IgG), directed against the obligatory N-methyl-D-aspartate-receptor subunit NR1 (NMDAR1-AB), belong to the mammalian autoimmune repertoire, and appear with age-dependently high seroprevalence across health and disease. Upon access to the brain, they can exert NMDAR-antagonistic/ketamine-like actions. Still unanswered key questions, addressed here, are conditions of NMDAR1-AB formation/boosting, intraindividual persistence/course in serum over time, and (patho)physiological significance of NMDAR1-AB in modulating neuropsychiatric phenotypes. We demonstrate in a translational fashion from mouse to human that (1) serum NMDAR1-AB fluctuate upon long-term observation, independent of blood-brain barrier (BBB) perturbation; (2) a standardized small brain lesion in juvenile mice leads to increased NMDAR1-AB seroprevalence (IgM + IgG), together with enhanced Ig-class diversity; (3) CTLA4 (immune-checkpoint) genotypes, previously found associated with autoimmune disease, predispose to serum NMDAR1-AB in humans; (4) finally, pursuing our prior findings of an early increase in NMDAR1-AB seroprevalence in human migrants, which implicated chronic life stress as inducer, we independently replicate these results with prospectively recruited refugee minors. Most importantly, we here provide the first experimental evidence in mice of chronic life stress promoting serum NMDAR1-AB (IgA). Strikingly, stress-induced depressive-like behavior in mice and depression/anxiety in humans are reduced in NMDAR1-AB carriers with compromised BBB where NMDAR1-AB can readily reach the brain. To conclude, NMDAR1-AB may have a role as endogenous NMDAR antagonists, formed or boosted under various circumstances, ranging from genetic predisposition to, e.g., tumors, infection, brain injury, and stress, altogether increasing over lifetime, and exerting a spectrum of possible effects, also including beneficial functions.


Assuntos
Autoanticorpos , Lesões Encefálicas , Animais , Barreira Hematoencefálica , Camundongos , Receptores de N-Metil-D-Aspartato , Estudos Soroepidemiológicos , Estresse Psicológico
20.
Semin Cell Dev Biol ; 98: 15-25, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31085323

RESUMO

Cancer is now considered a multifactorial disorder with different aetiologies and outcomes. Yet, all cancers share some common molecular features. Among these, the reprogramming of cellular metabolism has emerged as a key player in tumour initiation and progression. The finding that metabolic enzymes such as fumarate hydratase (FH), succinate dehydrogenase (SDH) and isocitrate dehydrogenase (IDH), when mutated, cause cancer suggested that metabolic dysregulation is not only a consequence of oncogenic transformation but that it can act as cancer driver. However, the mechanisms underpinning the link between metabolic dysregulation and cancer remain only partially understood. In this review we discuss the role of FH loss in tumorigenesis, focusing on the role of fumarate as a key activator of a variety of oncogenic cascades. We also discuss how these alterations are integrated and converge towards common biological processes. This review highlights the complexity of the signals elicited by FH loss, describes that fumarate can act as a bona fide oncogenic event, and provides a compelling hypothesis of the stepwise neoplastic progression after FH loss.


Assuntos
Fumarato Hidratase/metabolismo , Neoplasias/enzimologia , Fumarato Hidratase/genética , Humanos , Mutação , Neoplasias/metabolismo , Neoplasias/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA