Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 292
Filtrar
1.
Expert Opin Pharmacother ; 25(6): 685-694, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38717943

RESUMO

INTRODUCTION: Cytomegalovirus (CMV) remains a serious opportunistic infection in hematopoietic cell transplant (HCT) and solid-organ transplant (SOT) recipients. Traditional anti-CMV drugs are limited by toxicities and the development of resistance. Letermovir and maribavir are newly approved antivirals for the prevention and treatment of CMV. AREAS COVERED: Prior reviews have discussed use of letermovir for prevention of CMV after HCT and maribavir for resistant or refractory (R/R) CMV post HCT or SOT. Subsequent data have expanded their use including letermovir for primary CMV prophylaxis in high-risk renal transplant recipients and new recommendations for extending prophylaxis through day + 200 in certain HCT patients. Data on the use of maribavir for first asymptomatic CMV infection post-HCT has also been published. This review compares the pharmacology of anti-CMV agents and discusses the updated literature of these new drugs in the prevention and treatment of CMV. EXPERT OPINION: Letermovir and maribavir are much needed tools that spare toxicities of ganciclovir, foscarnet, and cidofovir. High cost is a challenge preventing their integration into clinical practice in resource-limited countries. Transplant centers need to exercise restraint in overuse to avoid resistance, particularly in the setting of high viral loads.


Assuntos
Acetatos , Antivirais , Infecções por Citomegalovirus , Farmacorresistência Viral , Transplante de Células-Tronco Hematopoéticas , Transplante de Órgãos , Quinazolinas , Ribonucleosídeos , Humanos , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/prevenção & controle , Antivirais/uso terapêutico , Antivirais/efeitos adversos , Antivirais/farmacologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Órgãos/efeitos adversos , Quinazolinas/uso terapêutico , Quinazolinas/farmacologia , Ribonucleosídeos/uso terapêutico , Ribonucleosídeos/farmacologia , Acetatos/uso terapêutico , Acetatos/efeitos adversos , Acetatos/farmacologia , Benzimidazóis/uso terapêutico , Benzimidazóis/efeitos adversos , Infecções Oportunistas/prevenção & controle , Infecções Oportunistas/tratamento farmacológico , Infecções Oportunistas/virologia , Carga Viral/efeitos dos fármacos , Diclororribofuranosilbenzimidazol/análogos & derivados
2.
J Pediatr Hematol Oncol ; 46(3): e244-e247, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38447094

RESUMO

Resistant and refractory cytomegalovirus (CMV) viremia can limit the provision of chemotherapy due to myelosuppression and end-organ dysfunction. Few therapies are available for children with clinically significant CMV viremia. We successfully used maribavir for a 4-year-old patient with lymphoma to complete his chemotherapy course. Resistance to maribavir did result after many months of therapy.


Assuntos
Infecções por Citomegalovirus , Diclororribofuranosilbenzimidazol , Neoplasias , Ribonucleosídeos , Pré-Escolar , Humanos , Antivirais/uso terapêutico , Benzimidazóis/uso terapêutico , Infecções por Citomegalovirus/tratamento farmacológico , Diclororribofuranosilbenzimidazol/análogos & derivados , Neoplasias/tratamento farmacológico , Ribonucleosídeos/uso terapêutico , Viremia/tratamento farmacológico
3.
Transpl Infect Dis ; 26(2): e14216, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38221739

RESUMO

BACKGROUND: Cytomegalovirus (CMV) infections among hematopoietic stem cell transplant (HSCT) and solid organ transplant (SOT) recipients impose a significant health care resource utilization (HCRU)-related economic burden. Maribavir (MBV), a novel anti-viral therapy (AVT), approved by the United States Food and Drug Administration for post-transplant CMV infections refractory (with/without resistance) to conventional AVTs has demonstrated lower hospital length of stay (LOS) versus investigator-assigned therapy (IAT; valgancilovir, ganciclovir, foscarnet, or cidofovir) in a phase 3 trial (SOLSTICE). This study estimated the HCRU costs of MBV versus IAT. METHODS: An economic model was developed to estimate HCRU costs for patients treated with MBV or IAT. Mean per-patient-per-year (PPPY) HCRU costs were calculated using (i) annualized mean hospital LOS in SOLSTICE, and (ii) CMV-related direct costs from published literature. Probabilistic sensitivity analysis with Monte-Carlo simulations assessed model robustness. RESULTS: Of 352 randomized patients receiving MBV (n = 235) or IAT (n = 117) for 8 weeks in SOLSTICE, 40% had HSCT and 60% had SOT. Mean overall PPPY HCRU costs of overall hospital-LOS were $67,205 (95% confidence interval [CI]: $33,767, $231,275) versus $145,501 (95% CI: $62,064, $589,505) for MBV and IAT groups, respectively. Mean PPPY ICU and non-ICU stay costs were: $32,231 (95% CI: $5,248, $184,524) versus $45,307 (95% CI: $3,957, $481,740) for MBV and IAT groups, and $82,237 (95% CI: $40,397, $156,945) MBV versus $228,329 (95% CI: $94,442, $517,476) for MBV and IAT groups, respectively. MBV demonstrated cost savings in over 99.99% of simulations. CONCLUSIONS: This analysis suggests that Mean PPPY HCRU costs were 29%-64% lower with MBV versus other-AVTs.


Assuntos
Infecções por Citomegalovirus , Diclororribofuranosilbenzimidazol/análogos & derivados , Transplante de Órgãos , Ribonucleosídeos , Humanos , Citomegalovirus , Antivirais , Ganciclovir/uso terapêutico , Hospitalização , Transplantados , Benzimidazóis/uso terapêutico , Ribonucleosídeos/uso terapêutico , Ribonucleosídeos/efeitos adversos , Transplante de Órgãos/efeitos adversos , Células-Tronco Hematopoéticas
4.
Clin Transl Sci ; 17(1): e13696, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38071422

RESUMO

Maribavir is an oral benzimidazole riboside for treatment of post-transplant cytomegalovirus (CMV) infection/disease that is refractory to prior antiviral treatment (with or without resistance). Through competitive inhibition of adenosine triphosphate, maribavir prevents the phosphorylation actions of UL97 to inhibit CMV DNA replication, encapsidation, and nuclear egress. Maribavir is active against CMV strains with viral DNA polymerase mutations that confer resistance to other CMV antivirals. After oral administration, maribavir is rapidly and highly absorbed (fraction absorbed >90%). The approved dose of 400 mg twice daily (b.i.d.) achieves a steady-state area under the curve per dosing interval of 128 h*µg/mL and trough concentration of 4.90 µg/mL (13.0 µM). Maribavir is highly bound to human plasma proteins (98%) with a small apparent volume of distribution of 27.3 L. Maribavir is primarily cleared by hepatic CYP3A4 metabolism; its major metabolite, VP44669 (pharmacologically inactive), is excreted in the urine and feces. There is no clinically relevant impact on maribavir pharmacokinetics by age, sex, race/ethnicity, body weight, transplant type, or hepatic/renal impairment status. In phase II dose-ranging studies, maribavir showed similar rates of CMV viral clearance across 400, 800, or 1200 mg b.i.d. groups, ranging from 62.5-70% in study 202 (NCT01611974) and 74-83% in study 203 (EudraCT 2010-024247-32). In the phase III SOLSTICE trial (NCT02931539), maribavir 400 mg b.i.d. demonstrated superior CMV viremia clearance at week 8 versus investigator-assigned treatments, with lower treatment discontinuation rates. Dysgeusia, nausea, vomiting, and diarrhea were commonly experienced adverse events among patients treated with maribavir in clinical trials.


Assuntos
Antivirais , Infecções por Citomegalovirus , Diclororribofuranosilbenzimidazol/análogos & derivados , Humanos , Ciência Translacional Biomédica , Infecções por Citomegalovirus/induzido quimicamente , Infecções por Citomegalovirus/tratamento farmacológico , Benzimidazóis
5.
Clin Infect Dis ; 78(3): 562-572, 2024 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-38036487

RESUMO

BACKGROUND: Neutropenia may limit the use of valganciclovir treatment for cytomegalovirus (CMV) infection following hematopoietic cell transplant (HCT). A phase 2 study indicated efficacy of maribavir with fewer treatment-limiting toxicities than valganciclovir. METHODS: In this multicenter, double-blind, phase 3 study, patients with first asymptomatic CMV infection post-HCT were stratified and randomized 1:1 to maribavir 400 mg twice daily or valganciclovir (dose-adjusted for renal clearance) for 8 weeks with 12 weeks of follow-up. The primary endpoint was confirmed CMV viremia clearance at week 8 (primary hypothesis of noninferiority margin of 7.0%). The key secondary endpoint was a composite of the primary endpoint with no findings of CMV tissue-invasive disease at week 8 through week 16. Treatment-emergent adverse events (TEAEs) were assessed. RESULTS: Among patients treated (273 maribavir; 274 valganciclovir), the primary endpoint of noninferiority of maribavir was not met (maribavir, 69.6%; valganciclovir, 77.4%; adjusted difference: -7.7%; 95% confidence interval [CI]: -14.98, -.36; lower limit of 95% CI of treatment difference exceeded -7.0%). At week 16, 52.7% and 48.5% of patients treated (maribavir and valganciclovir, respectively) maintained CMV viremia clearance without tissue-invasive disease (adjusted difference: 4.4%; 95% CI: -3.91, 12.76). With maribavir (vs valganciclovir), fewer patients experienced neutropenia (16.1% and 52.9%) or discontinued due to TEAEs (27.8% and 41.2%). Discontinuations were mostly due to neutropenia (maribavir, 4.0%; valganciclovir, 17.5%). CONCLUSIONS: Although noninferiority of maribavir to valganciclovir for the primary endpoint was not achieved based on the prespecified noninferiority margin, maribavir demonstrated comparable CMV viremia clearance during post-treatment follow-up, with fewer discontinuations due to neutropenia. Clinical Trials Registration. NCT02927067 [AURORA].


Assuntos
Infecções por Citomegalovirus , Diclororribofuranosilbenzimidazol , Transplante de Células-Tronco Hematopoéticas , Neutropenia , Humanos , Antivirais/efeitos adversos , Diclororribofuranosilbenzimidazol/análogos & derivados , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Neutropenia/induzido quimicamente , Valganciclovir/efeitos adversos , Viremia/tratamento farmacológico
6.
Sci Rep ; 13(1): 12621, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37537243

RESUMO

The effective treatment of breast cancer remains a profound clinical challenge, especially due to drug resistance and metastasis which unfortunately arise in many patients. The transcription inhibitor 5,6-dichloro-1-beta-D-ribofuranosyl-benzimidazole (DRB), as a selective inhibitor of cyclin-dependent kinase 9, was shown to be effective in inducing apoptosis in various hematopoietic malignancies. However, the anticancer efficacy of DRB against breast cancer is still unclear. Herein, we demonstrated that administration of DRB to the breast cancer cell line led to the inhibition of cellular proliferation and induction of the typical signs of apoptotic cells, including the increases in Annexin V-positive cells, DNA fragmentation, and activation of caspase-7, caspase-9, and poly (ADP ribose) polymerase (PARP). Treatment of DRB resulted in a rapid decline in the myeloid cell leukemia 1 (Mcl-1) protein, whereas levels of other antiapoptotic proteins did not change. Overexpression of Mcl-1 decreased the DRB-induced PARP cleavage, whereas knockdown of Mcl-1 enhanced the effects of DRB on PARP activation, indicating that loss of Mcl-1 accounts for the DRB-mediated apoptosis in MCF-7 cells, but not in T-47D. Furthermore, we found that co-treatment of MCF-7 cells with an inhibitor of AKT (LY294002) or an inhibitor of the proteasome (MG-132) significantly augmented the DRB-induced apoptosis. These data suggested that DRB in combination with LY294002 or MG-132 may have a greater therapeutic potency against breast cancer cells.


Assuntos
Neoplasias da Mama , Diclororribofuranosilbenzimidazol , Feminino , Humanos , Apoptose , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Diclororribofuranosilbenzimidazol/farmacologia
7.
Antimicrob Agents Chemother ; 66(9): e0240521, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-35916518

RESUMO

Maribavir was approved by the U.S. Food and Drug Administration in November 2021 for the treatment of adult and pediatric patients with post-transplant cytomegalovirus (CMV) infection/disease that is refractory to treatment (with or without genotypic resistance) with ganciclovir, valganciclovir, cidofovir, or foscarnet. Maribavir is an oral benzimidazole riboside with potent and selective multimodal anti-CMV activity. It utilizes a novel mechanism of action which confers activity against CMV strains that are resistant to traditional anti-CMV agents, and also offers a more favorable safety profile relative to the dose-limiting side effects of previously available therapies. Maribavir was initially studied as an agent for CMV prophylaxis in solid organ and hematopoietic stem cell recipients, but initial phase III trials failed to meet clinical efficacy endpoints. It has been more recently studied as a therapeutic agent at higher doses for refractory-resistant (R-R) CMV infections with favorable outcomes. After an overview of maribavir's chemistry and clinical pharmacology, this review will summarize clinical efficacy, safety, tolerability, and resistance data associated with maribavir therapy.


Assuntos
Anti-Infecciosos , Infecções por Citomegalovirus , Adulto , Anti-Infecciosos/farmacologia , Antivirais/farmacologia , Antivirais/uso terapêutico , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Criança , Cidofovir , Infecções por Citomegalovirus/prevenção & controle , Diclororribofuranosilbenzimidazol/análogos & derivados , Farmacorresistência Viral , Foscarnet , Ganciclovir/uso terapêutico , Humanos , Valganciclovir/uso terapêutico
8.
Clin Infect Dis ; 75(4): 690-701, 2022 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34864943

RESUMO

BACKGROUND: Therapies for refractory cytomegalovirus infections (with or without resistance [R/R]) in transplant recipients are limited by toxicities. Maribavir has multimodal anti-cytomegalovirus activity through the inhibition of UL97 protein kinase. METHODS: In this phase 3, open-label study, hematopoietic-cell and solid-organ transplant recipients with R/R cytomegalovirus were randomized 2:1 to maribavir 400 mg twice daily or investigator-assigned therapy (IAT; valganciclovir/ganciclovir, foscarnet, or cidofovir) for 8 weeks, with 12 weeks of follow-up. The primary endpoint was confirmed cytomegalovirus clearance at end of week 8. The key secondary endpoint was achievement of cytomegalovirus clearance and symptom control at end of week 8, maintained through week 16. RESULTS: 352 patients were randomized (235 maribavir; 117 IAT). Significantly more patients in the maribavir versus IAT group achieved the primary endpoint (55.7% vs 23.9%; adjusted difference [95% confidence interval (CI)]: 32.8% [22.80-42.74]; P < .001) and key secondary endpoint (18.7% vs 10.3%; adjusted difference [95% CI]: 9.5% [2.02-16.88]; P = .01). Rates of treatment-emergent adverse events (TEAEs) were similar between groups (maribavir, 97.4%; IAT, 91.4%). Maribavir was associated with less acute kidney injury versus foscarnet (8.5% vs 21.3%) and neutropenia versus valganciclovir/ganciclovir (9.4% vs 33.9%). Fewer patients discontinued treatment due to TEAEs with maribavir (13.2%) than IAT (31.9%). One patient per group had fatal treatment-related TEAEs. CONCLUSIONS: Maribavir was superior to IAT for cytomegalovirus viremia clearance and viremia clearance plus symptom control maintained post-therapy in transplant recipients with R/R cytomegalovirus. Maribavir had fewer treatment discontinuations due to TEAEs than IAT. Clinical Trials Registration. NCT02931539 (SOLSTICE).


Assuntos
Infecções por Citomegalovirus , Viremia , Antivirais/efeitos adversos , Citomegalovirus , Diclororribofuranosilbenzimidazol/análogos & derivados , Farmacorresistência Viral , Foscarnet/uso terapêutico , Ganciclovir/uso terapêutico , Humanos , Valganciclovir/uso terapêutico , Viremia/tratamento farmacológico
9.
Molecules ; 26(7)2021 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-33807474

RESUMO

Protein kinases are a large class of enzymes with numerous biological roles and many have been implicated in a vast array of diseases, including cancer and the novel coronavirus infection COVID-19. Thus, the development of chemical probes to selectively target each kinase is of great interest. Inhibition of protein kinases with ATP-competitive inhibitors has historically been the most widely used method. However, due to the highly conserved structures of ATP-sites, the identification of truly selective chemical probes is challenging. In this review, we use the Ser/Thr kinase CK2 as an example to highlight the historical challenges in effective and selective chemical probe development, alongside recent advances in the field and alternative strategies aiming to overcome these problems. The methods utilised for CK2 can be applied to an array of protein kinases to aid in the discovery of chemical probes to further understand each kinase's biology, with wide-reaching implications for drug development.


Assuntos
Caseína Quinase II/metabolismo , Sondas Moleculares/química , Inibidores de Proteínas Quinases/química , Proteínas Quinases/química , Proteínas Quinases/metabolismo , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , COVID-19 , Caseína Quinase II/química , Diclororribofuranosilbenzimidazol/química , Diclororribofuranosilbenzimidazol/farmacologia , Humanos , Sondas Moleculares/metabolismo , Naftiridinas/química , Naftiridinas/farmacologia , Fenazinas/química , Fenazinas/farmacologia , Polifenóis/química , Polifenóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia
10.
J Cell Physiol ; 234(5): 6023-6032, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30584668

RESUMO

Acute lung injury (ALI) is a life-threatening, diffuse heterogeneous lung injury characterized by acute onset, pulmonary edema, and respiratory failure. Lipopolysaccharide (LPS) is a leading cause for ALI and when administered to a mouse it induces a lung phenotype exhibiting some of the clinical characteristics of human ALI. This study focused on investigating whether microRNA-27b (miR-27b) affects ALI in a mouse model established by LPS-induction and to further explore the underlying mechanism. After model establishment, the mice were treated with miR-27b agomir, miR-27b antagomir, or D-ribofuranosylbenzimidazole (an inhibitor of nuclear factor-E2-related factor 2 [Nrf2]) to determine levels of miR-27b, Nrf2, nuclear factor kappa-light-chain-enhancer of activated B cells nuclear factor κB (NF-κB), p-NF-κB, and heme oxygenase-1 (HO-1). The levels of interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α (TNF-α) in bronchoalveolar lavage fluid (BALF) were determined. The results of luciferase activity suggested that Nrf2 was a target gene of miR-27b. It was indicated that the Nrf2 level decreased in lung tissues from ALI mice. The downregulation of miR-27b decreased the levels of IL-1ß, IL-6, and TNF-α in BALF of ALI mice. Downregulated miR-27b increased Nrf2 level, thus enhancing HO-1 level along with reduction of NF-κB level as well as the extent of NF-κB phosphorylation in the lung tissues of the transfected mice. Pathological changes were ameliorated in LPS-reduced mice elicited by miR-27b inhibition. The results of this study demonstrate that downregulated miR-27b couldenhance Nrf2 and HO-1 expressions, inhibit NF-κB signaling pathway, which exerts a protective effect on LPS-induced ALI in mice.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Antagomirs/farmacologia , Anti-Inflamatórios/farmacologia , Diclororribofuranosilbenzimidazol/farmacologia , Pulmão/efeitos dos fármacos , MicroRNAs/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , NF-kappa B/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Lipopolissacarídeos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/genética , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/metabolismo , Edema Pulmonar/patologia , Edema Pulmonar/prevenção & controle , Transdução de Sinais
11.
J Leukoc Biol ; 100(4): 699-709, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27154353

RESUMO

Retention of T cells within affected tissue is a critical component of adaptive immune inflammation. However, the mechanisms involved in T cell retention remain largely undefined. Previous studies revealed the capacity of cAMP signaling to regulate immune cell migration, as well as dynamic regulation of receptors that could induce cAMP production in immune cells. The potential for cAMP to act as a retention signal has been mostly unexplored, partially as a result of this second messenger's well-characterized inhibition of effector function in immune cells. Here, we report that cAMP regulates the tissue retention of mouse T cells at concentrations well below those that inhibited proliferation or decreased acquisition of an effector phenotype. Stimulation of CD4+ T cells with odorants known to be cognate ligands for T cell-expressed olfactory receptors induced cAMP and inhibited chemokine-driven chemotaxis without decreasing T cell proliferation or effector functions. Similar effects were observed following treatment with relatively low concentrations of the cAMP analog Sp-5,6-dichloro-1-ß-d-ribofuranosylbenzimidazole-3',5'-monophosphorothioate. Furthermore, pretreatment with odorants or cAMP at concentrations that did not inhibit effector function induced T cell tissue retention in mice by inhibiting chemokine-dependent T cell egress from the footpad to the draining lymph node. Together, these results suggest that odorant receptor-mediated increases in intracellular cAMP can modulate T cell tissue trafficking and may offer new therapeutic targets for controlling T cell tissue accumulation.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Quimiotaxia de Leucócito/efeitos dos fármacos , AMP Cíclico/biossíntese , Ácidos Dicarboxílicos/farmacologia , Odorantes , Imunidade Adaptativa , Animais , Animais Congênicos , Antígenos CD/biossíntese , Antígenos de Diferenciação de Linfócitos T/biossíntese , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/transplante , Linhagem Celular Tumoral , Células Cultivadas , Quimiocina CCL21/farmacologia , Quimiocina CXCL12/farmacologia , Colforsina/farmacologia , AMP Cíclico/farmacologia , Diclororribofuranosilbenzimidazol/análogos & derivados , Diclororribofuranosilbenzimidazol/farmacologia , Ácidos Graxos/farmacologia , Hidrazonas/farmacologia , Isoxazóis/farmacologia , Lectinas Tipo C/biossíntese , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Receptores Odorantes/sangue , Receptores Odorantes/efeitos dos fármacos , Tionucleotídeos/farmacologia
12.
Cell Rep ; 15(3): 611-624, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27068474

RESUMO

Steady-state circular RNAs (circRNAs) have been mapped to thousands of genomic loci in mammals. We studied circRNA processing using metabolic tagging of nascent RNAs with 4-thiouridine (4sU). Strikingly, the efficiency of circRNA processing from pre-mRNA is extremely low endogenously. Additional studies revealed that back-splicing outcomes correlate with fast RNA Polymerase II elongation rate and are tightly controlled by cis-elements in vivo. Additionally, prolonged 4sU labeling in cells shows that circRNAs are largely processed post-transcriptionally and that circRNAs are stable. Circular RNAs that are abundant at a steady-state level tend to accumulate. This is particularly true in cells, such as neurons, that have slow division rates. This study uncovers features of circRNA biogenesis by investigating the link between nascent circRNA processing and transcription.


Assuntos
RNA/biossíntese , Sequência de Bases , Linhagem Celular Tumoral , Diclororribofuranosilbenzimidazol/metabolismo , Humanos , Neurônios/metabolismo , Prosencéfalo/citologia , RNA Polimerase II/metabolismo , Splicing de RNA/genética , RNA Circular , Análise de Sequência de RNA , Spliceossomos/metabolismo , Tiouridina/metabolismo , Transcrição Gênica
13.
Wound Repair Regen ; 24(1): 89-99, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26646714

RESUMO

The goal of this study was to develop and validate a simple but quantitative cell-based assay to identify compounds that might be used pharmaceutically to give tissue repair a more regenerative character. The cornea was used as the model, and some specific aspects of repair in this organ were incorporated into assay design. A quantitative cell-based assay was developed based on transcriptional promoter activity of fibrotic marker genes ACT2A and TGFB2. Immortalized corneal stromal cells (HTK) or corneal epithelial cells (HCLE) were tested and compared to primary corneal stromal cells. Cells were transiently transfected with constructs containing the firefly luciferase reporter gene driven by transcriptional promoters for the selected fibrotic marker genes. A selected panel of seven chemical test compounds was used, containing three known fibrosis inhibitors: lovastatin (LOV), tyrphostin AG 1296 (6,7-dimethoxy-3-phenylquinoxaline) and SB203580 (4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole), and four potential fibrosis inhibitors: 5-iodotubercidin (4-amino-5-iodo-7-(ß-D-ribofuranosyl)-pyrrolo(2,3-d)pyrimidine), anisomycin, DRB (5,6-dichloro-1-ß-D-ribofuranosyl-benzimidazole) and latrunculin B. Transfected cells were treated with TGFB2 in the presence or absence of one of the test compounds. To validate the assay, compounds were tested for their direct effects on gene expression in the immortalized cell lines and primary human corneal keratocytes using RT-PCR and immunohistochemistry. Three "hits" were validated LOV, SB203580 and anisomycin. This assay, which can be applied in a high throughput format to screen large libraries of uncharacterized compounds, or known compounds that might be repurposed, offers a valuable tool for identifying new treatments to address a major unmet medical need. Anisomycin has not previously been characterized as antifibrotic, thus, this is a novel finding of the study.


Assuntos
Ceratócitos da Córnea/efeitos dos fármacos , Epitélio Corneano/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Actinas/efeitos dos fármacos , Actinas/genética , Animais , Anisomicina/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular , Córnea/citologia , Córnea/efeitos dos fármacos , Ceratócitos da Córnea/citologia , Técnicas Citológicas , Diclororribofuranosilbenzimidazol/farmacologia , Inibidores Enzimáticos/farmacologia , Epitélio Corneano/citologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Imidazóis/farmacologia , Lovastatina/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Piridinas/farmacologia , Coelhos , Tiazolidinas/farmacologia , Fator de Crescimento Transformador beta2/efeitos dos fármacos , Fator de Crescimento Transformador beta2/genética , Tubercidina/análogos & derivados , Tubercidina/farmacologia , Tirfostinas/farmacologia
14.
J Cell Biochem ; 117(1): 126-31, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26085436

RESUMO

In this work, we have found that casein kinase II (CKII) phosphorylates the CacyBP/SIP protein under in vitro conditions and have mapped the phosphorylation site to threonine 184. Moreover, we present evidence that S100A6, a CacyBP/SIP interacting protein, inhibits this phosphorylation in the presence of Ca(2+). CacyBP/SIP phosphorylation by CKII was also observed in neuroblastoma NB2a cells. Interestingly, we have found that the effect of DRB, a CKII inhibitor, on CacyBP/SIP phosphorylation state is similar to that of S100A6 overexpression. Phosphorylation at threonine 184 seems to have an effect on CacyBP/SIP phosphatase activity since the T184E phosphorylation mimic mutant overexpressed in NB2a cells has lower phosphatase activity toward p-ERK1/2 when compared to the non-phosphorylable T184A mutant or to the wild-type protein. In conclusion, our data suggest that S100A6 and Ca(2+), through inhibiting CacyBP/SIP phosphorylation on threonine 184, are important regulators of CacyBP/SIP phosphatase activity and of ERK1/2-Elk-1 signaling pathway.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas S100/metabolismo , Proteínas Elk-1 do Domínio ets/metabolismo , Animais , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Diclororribofuranosilbenzimidazol/farmacologia , Camundongos , Neuroblastoma/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteína A6 Ligante de Cálcio S100 , Proteínas S100/genética
15.
FEBS Lett ; 589(17): 2233-40, 2015 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-26183205

RESUMO

Immunomodulatory drugs such as thalidomide, lenalidomide, and pomalidomide exhibit high responsive rates for newly identified or relapsed multiple myeloma patients. However, their mechanisms of action are not completely understood. One mechanism involves the ubiquitination and degradation of two transcription factors, IKZF1 and IKZF3. Whether there are other degradation pathways for IKZF1 in myeloma cells remains unknown. Here, we found that although IKZF1 ubiquitination was reduced, its stability was also significantly reduced in MM1.S and OPM2 cells treated with kinase inhibitors, 5,6-dichlorobenzimidazole riboside (DRB) or roscovitine. Through pharmacological inhibition and biochemical approaches we demonstrated that instead of undergoing the ubiquitin-proteasome pathway, IKZF1 was degraded through apoptosis induced by kinase inhibition. This result may provide a new direction in developing therapeutic treatments for myeloma patients.


Assuntos
Apoptose/efeitos dos fármacos , Diclororribofuranosilbenzimidazol/farmacologia , Fator de Transcrição Ikaros/metabolismo , Purinas/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células HEK293 , Humanos , Immunoblotting , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Inibidores de Proteínas Quinases/farmacologia , Estabilidade Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Roscovitina , Fatores de Tempo , Ubiquitinação/efeitos dos fármacos
16.
Anticancer Res ; 34(12): 6981-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25503124

RESUMO

BACKGROUND: Positive transcription elongation factor-b (P-TEFb) is a complex containing CDK9 and a cyclin (T1, T2 or K). The effect of inhibition of P-TEFb by 5,6-dichloro-l-ß-D-ribofuranosyl benzimidazole (DRB) on cell radiosensitivity and the underlying mechanisms were investigated. MATERIALS AND METHODS: Six human cancer cell lines were subjected to (3)H-uridine incorporation, cell viability and clonogenic cell survival assays; cell-cycle redistribution and apoptosis assay; western blots and nuclear 53BP1 foci analysis after exposing the cells to DRB with/without γ-radiation. RESULTS: DRB suppressed colony formation and enhanced radiosensitivity of all cell lines. DRB caused a further increase in radiation-induced apoptosis and cell-cycle redistribution depending on p53 status. DRB prolonged the presence of radiation-induced nuclear p53 binding protein-1 (53BP1) foci and suppressed the expression of sirtuin-1 (SIRT1) and casein kinase 2-alpha (CK2α), suggesting an inhibition of DNA repair processes. CONCLUSION: Our findings indicate that DRB has the potential to increase the efficacy of radiotherapy and warrants further investigation using in vivo tumor models.


Assuntos
Caseína Quinase II/antagonistas & inibidores , Diclororribofuranosilbenzimidazol/farmacologia , Fator B de Elongação Transcricional Positiva/antagonistas & inibidores , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Sirtuína 1/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Caseína Quinase II/biossíntese , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos da radiação , Sirtuína 1/biossíntese , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
17.
Mol Cell ; 55(3): 347-60, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-24999238

RESUMO

Polycomb group (PcG) proteins are required for normal differentiation and development and are frequently deregulated in cancer. PcG proteins are involved in gene silencing; however, their role in initiation and maintenance of transcriptional repression is not well defined. Here, we show that knockout of the Polycomb repressive complex 2 (PRC2) does not lead to significant gene expression changes in mouse embryonic stem cells (mESCs) and that it is dispensable for initiating silencing of target genes during differentiation. Transcriptional inhibition in mESCs is sufficient to induce genome-wide ectopic PRC2 recruitment to endogenous PcG target genes found in other tissues. PRC2 binding analysis shows that it is restricted to nucleosome-free CpG islands (CGIs) of untranscribed genes. Our results show that it is the transcriptional state that governs PRC2 binding, and we propose that it binds by default to nontranscribed CGI genes to maintain their silenced state and to protect cell identity.


Assuntos
Ilhas de CpG , Células-Tronco Embrionárias/metabolismo , Inativação Gênica , Nucleossomos/genética , Nucleossomos/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Diclororribofuranosilbenzimidazol/farmacologia , Diterpenos/farmacologia , Epigênese Genética , Compostos de Epóxi/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Inativação de Genes , Inativação Gênica/efeitos dos fármacos , Genoma , Camundongos , Fenantrenos/farmacologia , Ligação Proteica/genética , Ligação Proteica/fisiologia
18.
Biochem Biophys Res Commun ; 449(3): 357-63, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24845383

RESUMO

In platelets, nitric oxide (NO) activates cGMP/PKG signalling, whereas prostaglandins and adenosine signal through cAMP/PKA. Cyclic nucleotide signalling has been considered to play an inhibitory role in platelets. However, an early stimulatory effect of NO and cGMP-PKG signalling in low dose agonist-induced platelet activation have recently been suggested. Here, we investigated whether different experimental conditions could explain some of the discrepancy reported for platelet cGMP-PKG-signalling. We treated gel-filtered human platelets with cGMP and cAMP analogues, and used flow cytometric assays to detect low dose thrombin-induced formation of small platelet aggregates, single platelet disappearance (SPD), platelet-derived microparticles (PMP) and thrombin receptor agonist peptide (TRAP)-induced P-selectin expression. All four agonist-induced platelet activation phases were blocked when platelets were costimulated with the PKG activators 8-Br-PET-cGMP or 8-pCPT-cGMP and low-doses of thrombin or TRAP. However, extended incubation with 8-Br-PET-cGMP decreased its inhibition of TRAP-induced P-selectin expression in a time-dependent manner. This effect did not involve desensitisation of PKG or PKA activity, measured as site-specific VASP phosphorylation. Moreover, PKG activators in combination with the PKA activator Sp-5,6-DCL-cBIMPS revealed additive inhibitory effect on TRAP-induced P-selectin expression. Taken together, we found no evidence for a stimulatory role of cGMP/PKG in platelets activation and conclude rather that cGMP/PKG signalling has an important inhibitory function in human platelet activation.


Assuntos
Micropartículas Derivadas de Células/efeitos dos fármacos , GMP Cíclico/análogos & derivados , Selectina-P/biossíntese , Agregação Plaquetária/efeitos dos fármacos , Receptores de Trombina/fisiologia , Tionucleotídeos/farmacologia , Trombina/farmacologia , Micropartículas Derivadas de Células/fisiologia , Células Cultivadas , AMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Diclororribofuranosilbenzimidazol/análogos & derivados , Diclororribofuranosilbenzimidazol/farmacologia , Humanos , Fatores de Tempo
19.
Histochem Cell Biol ; 141(3): 243-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24429832

RESUMO

A new method is proposed which combines the high spatial resolution of transmission electron microscopy with information on the dynamics of transcription. Incorporation of two different RNA precursors was used to define a time transcription window on cultured cells treated with hypometabolizing peptides which are known to modulate transcription. This procedure allows detecting a single fibril of newly synthesized RNA in the time range in which it is transcribed.


Assuntos
Leucina Encefalina-2-Alanina/farmacologia , Precursores de RNA/química , RNA Ribossômico/genética , Transcrição Gênica/genética , Linhagem Celular Tumoral , Núcleo Celular , Proliferação de Células , Dactinomicina/farmacologia , Diclororribofuranosilbenzimidazol/farmacologia , Leucina Encefalina-2-Alanina/análogos & derivados , Células HeLa , Humanos , Imuno-Histoquímica/métodos , Microscopia Eletrônica de Transmissão/métodos , Precursores de RNA/genética , RNA Ribossômico/biossíntese , Coloração e Rotulagem/métodos , Transcrição Gênica/efeitos dos fármacos , Uridina/análogos & derivados , Uridina/química
20.
Endocrine ; 47(1): 117-28, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24366643

RESUMO

This study aimed to assess the role of constitutive protein kinase CK2 in cytokine-induced activation of NFκB in pancreatic ß cell death. The CK2 inhibitors DRB (5,6-dichloro-1-ß-D-ribofuranosylbenzimidazole) (50 µM) and DMAT (2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole) (5 µM), which decreased CK2 activity by approx. 65 %, rescued INS-1E ß cells and mouse islets from cytokine (IL-1ß, TNF-α plus IFN-γ)-induced ß cell death without affecting H2O2- or palmitate-induced ß cell death. Western blot analysis revealed that while DRB or DMAT did not influence cytokine-induced IκBα degradation, they inhibited NFκB-dependent IκBα resynthesis, demonstrating that cytokine-induced NFκB activity is dependent on CK2. Both DRB and DMAT inhibited the constitutive phosphorylation of NFκB p65 at serine 529, while leaving cytokine-induced phosphorylations of NFκB p65 at serines 276 and 536 unaltered. In comparison, putative phosphorylation sites for CK2 on HDACs 1, 2, and 3 at serines 421/423, 394, and 424, respectively, which may stimulate NFκB transcriptional activity, were unchanged by cytokines and CK2 inhibitors. Whereas IL-1ß and TNF-α stimulate IκBα degradation and NFκB activation, IFN-γ potentiates cytokine-induced ß cell death through activation of STAT1. DRB and DMAT inhibited IFN-γ-stimulated phosphorylation of STAT1 at serine 727, while leaving IFN-γ-induced phosphorylation of STAT1 at tyrosine 701 unaffected. Inhibition of cytokine-induced ß cell death by CK2 inhibitors was, however, not dependent on IFN-γ, and IFN-γ did not affect CK2-dependent IκBα turnover. In conclusion, it is suggested that cytokine-induced activation of NFκB in ß cells is dependent on CK2 activity, which phosphorylates NFκB p65 at serine 529.


Assuntos
Caseína Quinase II/fisiologia , Citocinas/farmacologia , Células Secretoras de Insulina/fisiologia , NF-kappa B/metabolismo , Animais , Benzimidazóis/farmacologia , Caseína Quinase II/antagonistas & inibidores , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Células Cultivadas , Citocinas/metabolismo , Diclororribofuranosilbenzimidazol/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacologia , Masculino , Camundongos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA