Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 132(16)2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35763353

RESUMO

Targeted protein degradation is a rapidly advancing and expanding therapeutic approach. Drugs that degrade GSPT1 via the CRL4CRBN ubiquitin ligase are a new class of cancer therapy in active clinical development with evidence of activity against acute myeloid leukemia in early-phase trials. However, other than activation of the integrated stress response, the downstream effects of GSPT1 degradation leading to cell death are largely undefined, and no murine models are available to study these agents. We identified the domains of GSPT1 essential for cell survival and show that GSPT1 degradation leads to impaired translation termination, activation of the integrated stress response pathway, and TP53-independent cell death. CRISPR/Cas9 screens implicated decreased translation initiation as protective following GSPT1 degradation, suggesting that cells with higher levels of translation are more susceptible to the effects of GSPT1 degradation. We defined 2 Crbn amino acids that prevent Gspt1 degradation in mice, generated a knockin mouse with alteration of these residues, and demonstrated the efficacy of GSPT1-degrading drugs in vivo with relative sparing of numbers and function of long-term hematopoietic stem cells. Our results provide a mechanistic basis for the use of GSPT1 degraders for the treatment of cancer, including TP53-mutant acute myeloid leukemia.


Assuntos
Leucemia , Fatores de Terminação de Peptídeos , Animais , Morte Celular , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/metabolismo , Proteólise
2.
Cureus ; 13(2): e13582, 2021 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-33796425

RESUMO

Olfactory groove meningiomas are slow-growing tumors that manifest with headaches, changes in vision, and personality changes. The anatomic location of these tumors makes psychiatric disturbances more common early in the stage of tumors than focal neurological deficits. The case study here describes a unique instance of an undiagnosed giant olfactory groove meningioma in a young mother who was charged with a felony of aggravated child abuse for the death of her toddler daughter. The patient underwent gross tumor resection and radiation therapy, which halted the visual decline, resolved the frontal headaches, and the patient showed improved mood. In this patient, the insidious onset of personality changes without obvious focal neurologic deficits until late as well as a history of depression likely contributed to the delayed diagnosis. Failure to notice these initial behavioral manifestations in these patients allows for further psychiatric and cognitive decline, which can result in devastating social consequences.

3.
Br J Neurosurg ; : 1-4, 2021 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-33629613

RESUMO

BACKGROUND: Urothelial cell carcinoma (UCC), the most common cancer of the urinary system, rarely metastasizes to the brain. 1-3 More rare still is the subset of patients with urothelial carcinoma brain metastases whose UCC primary is first diagnosed at the same time as their CNS metastatic disease, with oncologic workup prompted by CNS clinical manifestations.4 Paraneoplastic optic neuropathy (PON) is likewise a rare clinical entity, which has not yet been described in association with UCC brain metastases. CASE DESCRIPTION: Herein, we present the sentinel case of UCC believed to be of endometrial origin in an 81 year old woman initially presenting with symptoms of fatigue, nausea, vertigo, and rapidly deteriorating vision over the course of 1 month. Visual deterioration prompted neuro imaging remarkable for multiple supratentorial and infratentorial metastases as well as likely neoplastic inflammatory involvement of the bilateral optic nerves. The patient underwent a right temporal open brain biopsy, with pathology findings consistent with UCC. Subsequent PET scanning demonstrated a heavy burden of disease including an FDG-avid uterine mass with local and distal extension of disease including bilateral hydroureteronephrosis with obstruction of the distal ureters. The patient and her family elected to pursue home hospice without further workup or intervention. CONCLUSIONS: While this is the first such case presented, it is possible that UCC of the uterine wall represents a particularly aggressive form of the disease more prone to presenting with CNS metastases and PON.

4.
Blood ; 134(2): 160-170, 2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-31043423

RESUMO

Pharmacologic agents that modulate ubiquitin ligase activity to induce protein degradation are a major new class of therapeutic agents, active in a number of hematologic malignancies. However, we currently have a limited understanding of the determinants of activity of these agents and how resistance develops. We developed and used a novel quantitative, targeted mass spectrometry (MS) assay to determine the relative activities, kinetics, and cell-type specificity of thalidomide and 4 analogs, all but 1 of which are in clinical use or clinical trials for hematologic malignancies. Thalidomide analogs bind the CRL4CRBN ubiquitin ligase and induce degradation of particular proteins, but each of the molecules studied has distinct patterns of substrate specificity that likely underlie the clinical activity and toxicities of each drug. Our results demonstrate that the activity of molecules that induce protein degradation depends on the strength of ligase-substrate interaction in the presence of drug, the levels of the ubiquitin ligase, and the expression level of competing substrates. These findings highlight a novel mechanism of resistance to this class of drugs mediated by competition between substrates for access to a limiting pool of the ubiquitin ligase. We demonstrate that increased expression of a nonessential substrate can lead to decreased degradation of other substrates that are critical for antineoplastic activity of the drug, resulting in drug resistance. These studies provide general rules that govern drug-dependent substrate degradation and key differences between thalidomide analog activity in vitro and in vivo.


Assuntos
Proteólise/efeitos dos fármacos , Talidomida/análogos & derivados , Talidomida/química , Talidomida/farmacologia , Ubiquitina-Proteína Ligases/química , Neoplasias Hematológicas/enzimologia , Humanos , Especificidade por Substrato , Ubiquitina-Proteína Ligases/efeitos dos fármacos
5.
Blood ; 132(14): 1535-1544, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30064974

RESUMO

Thalidomide and its derivatives, lenalidomide and pomalidomide, are clinically effective treatments for multiple myeloma and myelodysplastic syndrome with del(5q). These molecules lack activity in murine models, limiting investigation of their therapeutic activity or toxicity in vivo. Here, we report the development of a mouse model that is sensitive to thalidomide derivatives because of a single amino acid change in the direct target of thalidomide derivatives, cereblon (Crbn). In human cells, thalidomide and its analogs bind CRBN and recruit protein targets to the CRL4CRBN E3 ubiquitin ligase, resulting in their ubiquitination and subsequent degradation by the proteasome. We show that mice with a single I391V amino acid change in Crbn exhibit thalidomide-induced degradation of drug targets previously identified in human cells, including Ikaros (Ikzf1), Aiolos (Ikzf3), Zfp91, and casein kinase 1a1 (Ck1α), both in vitro and in vivo. We use the Crbn I391V model to demonstrate that the in vivo therapeutic activity of lenalidomide in del(5q) myelodysplastic syndrome can be explained by heterozygous expression of Ck1α in del(5q) cells. We found that lenalidomide acts on hematopoietic stem cells with heterozygous expression of Ck1α and inactivation of Trp53 causes lenalidomide resistance. We further demonstrate that Crbn I391V is sufficient to confer thalidomide-induced fetal loss in mice, capturing a major toxicity of this class of drugs. Further study of the Crbn I391V model will provide valuable insights into the in vivo efficacy and toxicity of this class of drugs.


Assuntos
Antineoplásicos/farmacologia , Lenalidomida/farmacologia , Síndromes Mielodisplásicas/tratamento farmacológico , Proteínas do Tecido Nervoso/genética , Mutação Puntual , Talidomida/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antineoplásicos/química , Caseína Quinase I/metabolismo , Modelos Animais de Doenças , Feminino , Hematopoese/efeitos dos fármacos , Lenalidomida/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Talidomida/análogos & derivados
6.
Blood ; 132(11): 1095-1105, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-29954749

RESUMO

Truncating mutations in the terminal exon of protein phosphatase Mg2+/Mn2+ 1D (PPM1D) have been identified in clonal hematopoiesis and myeloid neoplasms, with a striking enrichment in patients previously exposed to chemotherapy. In this study, we demonstrate that truncating PPM1D mutations confer a chemoresistance phenotype, resulting in the selective expansion of PPM1D-mutant hematopoietic cells in the presence of chemotherapy in vitro and in vivo. Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein-9 nuclease mutational profiling of PPM1D in the presence of chemotherapy selected for the same exon 6 mutations identified in patient samples. These exon 6 mutations encode for a truncated protein that displays elevated expression and activity due to loss of a C-terminal degradation domain. Global phosphoproteomic profiling revealed altered phosphorylation of target proteins in the presence of the mutation, highlighting multiple pathways including the DNA damage response (DDR). In the presence of chemotherapy, PPM1D-mutant cells have an abrogated DDR resulting in altered cell cycle progression, decreased apoptosis, and reduced mitochondrial priming. We demonstrate that treatment with an allosteric, small molecule inhibitor of PPM1D reverts the phosphoproteomic, DDR, apoptotic, and mitochondrial priming changes observed in PPM1D-mutant cells. Finally, we show that the inhibitor preferentially kills PPM1D-mutant cells, sensitizes the cells to chemotherapy, and reverses the chemoresistance phenotype. These results provide an explanation for the enrichment of truncating PPM1D mutations in the blood of patients exposed to chemotherapy and in therapy-related myeloid neoplasms, and demonstrate that PPM1D can be a targeted in the prevention of clonal expansion of PPM1D-mutant cells and the treatment of PPM1D-mutant disease.


Assuntos
Sequência de Bases , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Neoplasias Hematológicas , Células-Tronco Hematopoéticas/enzimologia , Transtornos Mieloproliferativos , Proteínas de Neoplasias , Células-Tronco Neoplásicas/enzimologia , Proteína Fosfatase 2C , Deleção de Sequência , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/enzimologia , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patologia , Células-Tronco Hematopoéticas/patologia , Humanos , Transtornos Mieloproliferativos/tratamento farmacológico , Transtornos Mieloproliferativos/enzimologia , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/patologia , Proteína Fosfatase 2C/antagonistas & inibidores , Proteína Fosfatase 2C/genética , Proteína Fosfatase 2C/metabolismo
7.
Sci Transl Med ; 10(436)2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29643232

RESUMO

Thrombosis is a major cause of morbidity and mortality in Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), clonal disorders of hematopoiesis characterized by activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling. Neutrophil extracellular trap (NET) formation, a component of innate immunity, has been linked to thrombosis. We demonstrate that neutrophils from patients with MPNs are primed for NET formation, an effect blunted by pharmacological inhibition of JAK signaling. Mice with conditional knock-in of Jak2V617F, the most common molecular driver of MPN, have an increased propensity for NET formation and thrombosis. Inhibition of JAK-STAT signaling with the clinically available JAK2 inhibitor ruxolitinib abrogated NET formation and reduced thrombosis in a deep vein stenosis murine model. We further show that expression of PAD4, a protein required for NET formation, is increased in JAK2V617F-expressing neutrophils and that PAD4 is required for Jak2V617F-driven NET formation and thrombosis in vivo. Finally, in a population study of more than 10,000 individuals without a known myeloid disorder, JAK2V617F-positive clonal hematopoiesis was associated with an increased incidence of thrombosis. In aggregate, our results link JAK2V617F expression to NET formation and thrombosis and suggest that JAK2 inhibition may reduce thrombosis in MPNs through cell-intrinsic effects on neutrophil function.


Assuntos
Armadilhas Extracelulares/metabolismo , Neoplasias Hematológicas/metabolismo , Transtornos Mieloproliferativos/metabolismo , Trombose/metabolismo , Animais , Estudos de Casos e Controles , Proliferação de Células/fisiologia , Feminino , Neoplasias Hematológicas/tratamento farmacológico , Hidrolases/metabolismo , Janus Quinase 2/metabolismo , Janus Quinases/antagonistas & inibidores , Janus Quinases/metabolismo , Camundongos , Transtornos Mieloproliferativos/tratamento farmacológico , Nitrilas , Proteína-Arginina Desiminase do Tipo 4 , Pirazóis/uso terapêutico , Pirimidinas , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/fisiologia , Trombose/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA