Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 465
Filtrar
1.
Exp Neurol ; 372: 114612, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37993080

RESUMO

Edema formation is one of the very first events to occur after spinal cord injury (SCI) leading to an increase of the intrathecal pressure and consequently to serious spinal tissue and functional impairments. Current edema treatments are still symptomatic and/or non-specific. Since edema formation mechanisms are mainly described as vasogenic and cytotoxic, it becomes crucial to understand the interplay between these two subtypes. Acting on key targets to inhibit edema formation may reduce secondary damage and related functional impairments. In this study, we characterize the edema kinetic after T9-10 spinal contusion. We use trifluoperazine (TFP) to block the expression and the functional subcellular localization of aquaporin-4 supposed to be implicated in the cytotoxic edema formation. We also use sodium cromoglycate (SCG) to deactivate mast cell degranulation known to be implicated in the vasogenic edema formation. Our results show a significant reduction of edema after TFP treatment and after TFP-SCG combined treatment compared to control. This reduction is correlated with limited onset of initial sensorimotor impairments particularly after combined treatment. Our results highlight the importance of potential synergetic targets in early edema therapy after SCI as part of tissue sparing strategies.


Assuntos
Traumatismos da Medula Espinal , Medula Espinal , Ratos , Animais , Medula Espinal/metabolismo , Cromolina Sódica/farmacologia , Cromolina Sódica/uso terapêutico , Cromolina Sódica/metabolismo , Trifluoperazina/farmacologia , Trifluoperazina/uso terapêutico , Trifluoperazina/metabolismo , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo , Edema/tratamento farmacológico , Edema/etiologia
2.
Sci Adv ; 9(43): eadf1332, 2023 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-37878712

RESUMO

Cancers in the central nervous system resist therapies effective in other cancers, possibly due to the unique biochemistry of the human brain microenvironment composed of cerebrospinal fluid (CSF). However, the impact of CSF on cancer cells and therapeutic efficacy is unknown. Here, we examined the effect of human CSF on glioblastoma (GBM) tumors from 25 patients. We found that CSF induces tumor cell plasticity and resistance to standard GBM treatments (temozolomide and irradiation). We identified nuclear protein 1 (NUPR1), a transcription factor hampering ferroptosis, as a mediator of therapeutic resistance in CSF. NUPR1 inhibition with a repurposed antipsychotic, trifluoperazine, enhanced the killing of GBM cells resistant to chemoradiation in CSF. The same chemo-effective doses of trifluoperazine were safe for human neurons and astrocytes derived from pluripotent stem cells. These findings reveal that chemoradiation efficacy decreases in human CSF and suggest that combining trifluoperazine with standard care may improve the survival of patients with GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/metabolismo , Trifluoperazina/farmacologia , Trifluoperazina/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Temozolomida/farmacologia , Quimiorradioterapia , Linhagem Celular Tumoral , Microambiente Tumoral
3.
Int J Med Sci ; 20(6): 797-809, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37213674

RESUMO

Currently, no specific and standard treatment for traumatic brain injury (TBI) has been developed. Therefore, studies on new therapeutic drugs for TBI treatment are urgently needed. Trifluoperazine (TFP) is a therapeutic agent for the treatment of psychiatric disorders that reduces edema of the central nervous system. However, the specific working mechanism of TFP is not fully understood in TBI. In this study, the immunofluorescence co-localization analysis revealed that the area and intensity covered by Aquaporin4 (AQP4) on the surface of brain cells (astrocyte endfeet) increased significantly after TBI. In contrast, TFP treatment reversed these phenomena. This finding showed that TFP inhibited AQP4 accumulation on the surface of brain cells (astrocyte endfeet). The tunel fluorescence intensity and fluorescence area were lower in the TBI+TFP group compared to the TBI group. Additionally, the brain edema, brain defect area, and modified neurological severity score (mNSS) were lower in the TBI+TFP. The RNA-seq was performed on the cortical tissues of rats in the Sham, TBI, and TBI+TFP groups. A total of 3774 genes differently expressed between the TBI and the Sham group were identified. Of these, 2940 genes were up-regulated and 834 genes were down-regulated. A total of 1845 differently expressed genes between the TBI+TFP and TBI group were also identified, in which 621 genes were up-regulated and 1224 genes were down-regulated. Analysis of the common differential genes in the three groups showed that TFP could reverse the expression of apoptosis and inflammation genes. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that the differentially expressed genes (DEGs) were highly enriched in the signaling pathways regulating inflammation. In conclusion, TFP alleviates brain edema after TBI by preventing the accumulation of AQP4 on the surface of brain cells. Generally, TFP alleviates apoptosis and inflammatory response induced by TBI, and promotes the recovery of nerve function in rats after TBI. Thus, TFP is a potential therapeutic agent for TBI treatment.


Assuntos
Edema Encefálico , Lesões Encefálicas Traumáticas , Animais , Ratos , Apoptose/genética , Aquaporina 4/antagonistas & inibidores , Aquaporina 4/genética , Aquaporina 4/metabolismo , Encéfalo , Edema Encefálico/etiologia , Edema Encefálico/genética , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/genética , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/metabolismo , Trifluoperazina/farmacologia , Trifluoperazina/uso terapêutico , Trifluoperazina/metabolismo
4.
Head Neck ; 45(2): 316-328, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36349408

RESUMO

BACKGROUND: Trifluoperazine (TFP) is a typical antipsychotic primarily used to treat schizophrenia. In this study, we aimed to evaluate whether TFP can be used as a therapeutic agent against nasopharyngeal carcinoma (NPC) and identify its underlying molecular mechanisms. METHODS: We used NPC-TW01, TW03, TW04, and BM to assess the anticancer effects of TFP by using cytotoxicity, wound healing, colony formation, and cell invasion assays. An in vivo animal study was conducted. RNA sequencing combined with Ingenuity Pathways Analysis was performed to identify the mechanism by which TFP influences NPC cells. RESULTS: Our data revealed that TFP decreased NPC cell viability in a dose-dependent manner. The invasion and migration of NPC tumor cells were inhibited by TFP. An in vivo study also demonstrated the anticancer effects of TFP. RNA sequencing revealed several anticancer molecular mechanisms following TFP administration. CONCLUSIONS: The antipsychotic drug TFP could be a potential therapeutic regimen for NPC treatment.


Assuntos
Antipsicóticos , Neoplasias Nasofaríngeas , Animais , Antipsicóticos/farmacologia , Antipsicóticos/uso terapêutico , Trifluoperazina/farmacologia , Trifluoperazina/uso terapêutico , Carcinoma Nasofaríngeo/tratamento farmacológico , Linhagem Celular Tumoral , Neoplasias Nasofaríngeas/tratamento farmacológico , Proliferação de Células , Movimento Celular
5.
Arch Virol ; 167(11): 2203-2212, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35920983

RESUMO

Dengue virus (DENV), a member of the genus Flavivirus, family Flaviviridae, is the most widespread viral pathogen transmitted to humans by mosquitoes. Despite the increased incidence of DENV infection, there are no antiviral drugs available for treatment or prevention. Phenothiazines are heterocyclic compounds with various pharmacological properties that are very adaptable for drug repurposing. In the present report, we analyzed the antiviral activity against DENV and the related Zika virus (ZIKV) of trifluoperazine (TFP), a phenothiazine derivative in clinical use as an antipsychotic and antiemetic agent. TFP exhibited dose-dependent inhibitory activity against the four DENV serotypes and ZIKV in monkey Vero cells at non-cytotoxic concentrations with 50% effective concentration values in the range 1.6-6.4 µM. A similar level of antiviral efficacy was exhibited by TFP against flavivirus infection in the human cell lines A549 and HepG2. Mechanistic studies, performed using time-dependent infectivity assays, real-time RT-PCR, Western blot, and immunofluorescence techniques, indicated that uncoating of the virus during penetration into the cell was the main target for TFP in infected cells, but the compound also exerted a minor effect on a late stage of the virus multiplication cycle. This study demonstrates that TFP, a pharmacologically active phenothiazine, is a selective inhibitor of DENV multiplication in cell culture. Our findings open perspectives for the repositioning of phenothiazines like TFP with a wide spectrum of antiviral efficacy as potential agents for the control of pathogenic flaviviruses.


Assuntos
Antieméticos , Antipsicóticos , Vírus da Dengue , Dengue , Infecção por Zika virus , Zika virus , Animais , Antieméticos/farmacologia , Antieméticos/uso terapêutico , Antipsicóticos/farmacologia , Antipsicóticos/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico , Chlorocebus aethiops , Dengue/tratamento farmacológico , Humanos , Fenotiazinas/farmacologia , Fenotiazinas/uso terapêutico , Trifluoperazina/farmacologia , Trifluoperazina/uso terapêutico , Células Vero , Replicação Viral
6.
Tohoku J Exp Med ; 257(4): 315-326, 2022 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-35644544

RESUMO

Multiple myeloma (MM) is a common hematological malignancy. Bortezomib (BTZ) is a traditional medicine for MM treatment, but there are limitations for current treatment methods. Trifluoperazine (TFP) is a clinical drug for acute and chronic psychosis therapy. Lately, researchers have found that TFP can suppress tumor growth in many cancers. We attempted to study the effects of BTZ and TFP on MM in vivo and in vitro. We concentrated on the individual and combined impact of BTZ and TFP on the proliferation and apoptosis of MM cells via Cell Counting kit-8 assay, EdU assay, western blot, and flow cytometry. We found that combination therapy has a strong synergistic impact on MM cells. Combination therapy could induce cell arrest during G2/M phase and induce apoptosis in MM cells. Meanwhile, BTZ combined with TFP could play a better role in the anti-MM effect in vivo through MM.1s xenograft tumor models. Furthermore, we explored the mechanism of TFP-induced apoptosis in MM, and we noticed that TFP might induce MM apoptosis by inhibiting p-P38 MAPK/NUPR1. In summary, our findings suggest that TFP could synergistically enhance the BTZ-induced anti-cancer effect in multiple myeloma, which might be a promising therapeutic strategy for MM treatment.


Assuntos
Antineoplásicos , Mieloma Múltiplo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Mieloma Múltiplo/tratamento farmacológico , Proteínas de Neoplasias/metabolismo , Trifluoperazina/farmacologia , Trifluoperazina/uso terapêutico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Eur J Pharmacol ; 909: 174432, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34416238

RESUMO

Multiple sclerosis (MS) is one of the most common neurodegenerative diseases. In this disease, the immune system attacks oligodendrocyte cells and the myelin sheath of myelinated neurons in the central nervous system, causing their destruction. These conditions lead to impaired conduction of nerve impulses and are manifested by symptoms such as weakness, fatigue, visual and motor disorders. This study aimed to evaluate the ability of trifluoperazine (TF) to improve cuprizone-induced behavioral and histopathological changes in the prefrontal cortex of C57BL/6 male mice. Demyelination was induced by adding 0.2% cuprizone (CPZ) to the standard animal diet for 6 weeks. Three doses of TF (0.5, 1 and 2 mg/kg/day; i.p.) were given once daily for the last 2 weeks of treatment. Treatment with CPZ induced a weight loss during 6 weeks of treatment compared to the control group, which was reversed by the administration of TF. Behavioral tests (pole test and rotarod performance test) showed a decrease in motor coordination and balance in the group treated with CPZ (P < 0.01). Treatment with TF during the last two weeks was able to improve these motor deficiencies. Histopathological examination also evidenced an increase in demyelination in the CPZ group, which was improved by TF administration. In addition, CPZ intake significantly decreased the cerebral cortex levels of p-Nrf2 (P < 0.001) and increased the levels of p-IKB (P < 0.001) and, these changes were normalized in the TF groups. TF administration also reversed the increased levels of nitrite and the reduced activity of the antioxidant enzyme superoxide dismutase associated with CPZ exposure. TF can to reduce the harmful effects of CPZ by reducing the demyelination and modulating the Nrf2 and NF-kB signaling pathways.


Assuntos
Esclerose Múltipla/tratamento farmacológico , Bainha de Mielina/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Trifluoperazina/farmacologia , Animais , Cuprizona/administração & dosagem , Cuprizona/toxicidade , Modelos Animais de Doenças , Humanos , Proteínas I-kappa B/metabolismo , Masculino , Camundongos , Esclerose Múltipla/induzido quimicamente , Esclerose Múltipla/patologia , Bainha de Mielina/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Transdução de Sinais/efeitos dos fármacos , Trifluoperazina/uso terapêutico
9.
Biochim Biophys Acta Biomembr ; 1863(5): 183573, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33561476

RESUMO

Stroke is the second leading cause of death and the third leading cause of disability globally. Edema is a hallmark of stroke resulting from dysregulation of water homeostasis in the central nervous system (CNS) and plays the major role in stroke-associated morbidity and mortality. The overlap between cellular and vasogenic edema makes treating this condition complicated, and to date, there is no pathogenically oriented drug treatment for edema. Water balance in the brain is tightly regulated, primarily by aquaporin 4 (AQP4) channels, which are mainly expressed in perivascular astrocytic end-feet. Targeting AQP4 could be a useful therapeutic approach for treating brain edema; however, there is no approved drug for stroke treatment that can directly block AQP4. In this study, we demonstrate that the FDA-approved drug trifluoperazine (TFP) effectively reduces cerebral edema during the early acute phase in post-stroke mice using a photothrombotic stroke model. This effect was combined with an inhibition of AQP4 expression at gene and protein levels. Importantly, TFP does not appear to induce any deleterious changes on brain electrolytes or metabolic markers, including total protein or lipid levels. Our results support a possible role for TFP in providing a beneficial extra-osmotic effect on brain energy metabolism, as indicated by the increase of glycogen levels. We propose that targeting AQP4-mediated brain edema using TFP is a viable therapeutic strategy during the early and acute phase of stroke that can be further investigated during later stages to help in developing novel CNS edema therapies.


Assuntos
Aquaporina 4/metabolismo , Biomarcadores/metabolismo , Encéfalo/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Trifluoperazina/uso terapêutico , Animais , Aquaporina 4/genética , Modelos Animais de Doenças , Glicogênio/química , Glicogênio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Agregados Proteicos , Acidente Vascular Cerebral/metabolismo
10.
Pharmacol Res ; 163: 105295, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33176207

RESUMO

Targeted therapies and immunotherapy have brought substantial benefits to patients with melanoma. However, brain metastases remain the biggest threat to the survival and quality of life of melanoma patients. One of the major challenges to an effective therapy is the inability of drugs to penetrate the blood-brain barrier (BBB). Anti-schizophrenic drugs can cross the BBB, and many of them have demonstrated anti-cancer effects. Repurposing existing drugs for new clinical indications is an alluring strategy for anticancer drug discovery. Herein, we applied this strategy and screened a small collection of existing anti-schizophrenic drugs to use as anti-melanoma agents. Among them, trifluoperazine dihydrochloride (TFP) exhibited promising potencies for suppressing the growth and metastasis of melanoma, both in vitro and in vivo. TFP obviously suppressed the viability of melanoma cells within the micromolar range and inhibited the growth of melanoma in the subcutaneous mice models. Notably, intraperitoneal (i.p.) administration of TFP (40 mg/kg/day) obviously inhibited the growth of intra-carotid-injection established melanoma brain metastasis and extended the survival of brain metastasis-bearing mice. Moreover, TFP significantly suppressed lung metastasis and bone metastasis of melanoma in preclinical metastasis models. Mechanistically, TFP caused G0/G1 cell cycle arrest and mitochondrial-dependent intrinsic apoptosis of melanoma cells. In addition, TFP treatment increased the expression of microtubule associated protein 1 light chain 3 beta-II (LC3B-II) and p62 in vitro, suggesting an inhibition of autophagic flux. TFP decreased LysoTracker Red uptake after treatment, indicating impaired acidification of lysosomes. Moreover, the colocalization of LC3 with lysosomal-associated membrane protein 1 (LAMP1), a lysosome marker, was also suppressed after TFP treatment, suggesting that TFP might block the fusion of autophagosomes with lysosomes, which led to autophagosome accumulation. Taken together, our data highlight the potential of repurposing TFP as a new adjuvant drug for treating melanoma patients with brain, lung, and bone metastases.


Assuntos
Antineoplásicos/uso terapêutico , Antipsicóticos/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Trifluoperazina/uso terapêutico , Animais , Autofagia/efeitos dos fármacos , Neoplasias Ósseas/secundário , Neoplasias Encefálicas/secundário , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Reposicionamento de Medicamentos , Feminino , Humanos , Neoplasias Pulmonares/secundário , Melanoma/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Neoplasias Cutâneas/patologia
11.
Proc Natl Acad Sci U S A ; 117(20): 11085-11096, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32358191

RESUMO

Glioblastoma (GBM) is the deadliest adult brain cancer, and all patients ultimately succumb to the disease. Radiation therapy (RT) provides survival benefit of 6 mo over surgery alone, but these results have not improved in decades. We report that radiation induces a glioma-initiating cell phenotype, and we have identified trifluoperazine (TFP) as a compound that interferes with this phenotype conversion. TFP causes loss of radiation-induced Nanog mRNA expression, and activation of GSK3 with consecutive posttranslational reduction in p-Akt, Sox2, and ß-catenin protein levels. TFP did not alter the intrinsic radiation sensitivity of glioma-initiating cells (GICs). Continuous treatment with TFP and a single dose of radiation reduced the number of GICs in vivo and prolonged survival in syngeneic and patient-derived orthotopic xenograft (PDOX) mouse models of GBM. Our findings suggest that the combination of a dopamine receptor antagonist with radiation enhances the efficacy of RT in GBM by preventing radiation-induced phenotype conversion of radiosensitive non-GICs into treatment-resistant, induced GICs (iGICs).


Assuntos
Antagonistas de Dopamina/farmacologia , Glioblastoma/metabolismo , Fenótipo , Receptores Dopaminérgicos/efeitos dos fármacos , Trifluoperazina/farmacologia , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Modelos Animais de Doenças , Antagonistas de Dopamina/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/radioterapia , Glioma/tratamento farmacológico , Glioma/metabolismo , Glioma/patologia , Glioma/radioterapia , Quinase 3 da Glicogênio Sintase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , RNA Mensageiro/metabolismo , Tolerância a Radiação , Fatores de Transcrição SOXB1 , Trifluoperazina/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina
12.
Drug Res (Stuttg) ; 70(6): 265-272, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32365382

RESUMO

The link between Ca2+ dysregulation, mitochondria damages, oxidative stress and cellular derangement is particularly evident in neurotoxicity induced by chemotherapeutic agents. In the current study, we investigated effects of trifluoperazine (TFP) as an inhibitor of calmodulin against the cytotoxicity induced by cytarabine (Ara-C) and Ifosfamide (IFOS) on isolated rat neurons and also the mechanisms involved in this toxicity. Isolated rat neurons were pretreated with TFP (100 µM) for 5 min at 37°C, then Ara-C (226 µM) and IFOS (290 µM) were added in separate experiments. After 3 h, the cytotoxicity, reactive oxygen species (ROS), lysosomal membrane destabilization, mitochondrial membrane potential (MMP), lipid peroxidation (LP), glutathione (GSH) and glutathione disulfide (GSSG) levels were measured. Ara-C and IFOS treatments caused a significant decrease in cellular viability, which was accompanied by ROS generation, GSSG/GSH ratio, lipid peroxidation and lysosomal and mitochondrial damages. On the other hand, TFP (100 µM) pre-treatment attenuated Ara-C and IFOS -induced decrease in cell viability. In addition, TFP (100 µM) pre-treatment significantly protected against Ara-C and IFOS -induced increase in ROS generation, lysosomal and mitochondrial damages, lipid peroxidation levels and decrease in GSH/GSSG ratio. Our data provided insights into the mechanism of protection by TFP against Ara-C and IFOS neurotoxicity, which is related, to neuronal ROS formation and mitochondrial damages.


Assuntos
Antineoplásicos/toxicidade , Mitocôndrias/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/prevenção & controle , Trifluoperazina/farmacologia , Animais , Encéfalo/citologia , Células Cultivadas , Citarabina/toxicidade , Modelos Animais de Doenças , Humanos , Ifosfamida/toxicidade , Peroxidação de Lipídeos/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/patologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Síndromes Neurotóxicas/etiologia , Estresse Oxidativo/efeitos dos fármacos , Cultura Primária de Células , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Trifluoperazina/uso terapêutico
13.
mBio ; 11(2)2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32156806

RESUMO

Frequent and excessive use of antibiotics primes patients to Clostridioides difficile infection (CDI), which leads to fatal pseudomembranous colitis, with limited treatment options. In earlier reports, we used a drug repurposing strategy and identified amoxapine (an antidepressant), doxapram (a breathing stimulant), and trifluoperazine (an antipsychotic), which provided significant protection to mice against lethal infections with several pathogens, including C. difficile However, the mechanisms of action of these drugs were not known. Here, we provide evidence that all three drugs offered protection against experimental CDI by reducing bacterial burden and toxin levels, although the drugs were neither bacteriostatic nor bactericidal in nature and had minimal impact on the composition of the microbiota. Drug-mediated protection was dependent on the presence of the microbiota, implicating its role in evoking host defenses that promoted protective immunity. By utilizing transcriptome sequencing (RNA-seq), we identified that each drug increased expression of several innate immune response-related genes, including those involved in the recruitment of neutrophils, the production of interleukin 33 (IL-33), and the IL-22 signaling pathway. The RNA-seq data on selected genes were confirmed by quantitative real-time PCR (qRT-PCR) and protein assays. Focusing on amoxapine, which had the best anti-CDI outcome, we demonstrated that neutralization of IL-33 or depletion of neutrophils resulted in loss of drug efficacy. Overall, our lead drugs promote disease alleviation and survival in the murine model through activation of IL-33 and by clearing the pathogen through host defense mechanisms that critically include an early influx of neutrophils.IMPORTANCEClostridioides difficile is a spore-forming anaerobic bacterium and the leading cause of antibiotic-associated colitis. With few therapeutic options and high rates of disease recurrence, the need to develop new treatment options is urgent. Prior studies utilizing a repurposing approach identified three nonantibiotic Food and Drug Administration-approved drugs, amoxapine, doxapram, and trifluoperazine, with efficacy against a broad range of human pathogens; however, the protective mechanisms remained unknown. Here, we identified mechanisms leading to drug efficacy in a murine model of lethal C. difficile infection (CDI), advancing our understanding of the role of these drugs in infectious disease pathogenesis that center on host immune responses to C. difficile Overall, these studies highlight the crucial involvement of innate immune responses, as well as the importance of immunomodulation as a potential therapeutic option to combat CDI.


Assuntos
Amoxapina/uso terapêutico , Infecções por Clostridium/tratamento farmacológico , Doxapram/uso terapêutico , Imunidade Inata , Microbiota/efeitos dos fármacos , Trifluoperazina/uso terapêutico , Animais , Clostridioides difficile/efeitos dos fármacos , Reposicionamento de Medicamentos , Feminino , Imunomodulação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/imunologia , RNA-Seq , Organismos Livres de Patógenos Específicos
14.
Int J Mol Sci ; 20(13)2019 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-31262032

RESUMO

Cisplatin-based chemotherapy is the primary treatment for metastatic bladder urothelial carcinoma (UC). Most patients inevitably encounter drug resistance and resultant disease relapse. Reduced apoptosis plays a critical role in chemoresistance. Trifluoperazine (TFP), an antipsychotic agent, has demonstrated antitumor effects on various cancers. This study investigated the efficacy of TFP in inhibiting cisplatin-resistant bladder UC and explored the underlying mechanism. Our results revealed that cisplatin-resistant UC cells (T24/R) upregulated the antiapoptotic factor, B-cell lymphoma-extra large (Bcl-xL). Knockdown of Bcl-xL by siRNA resensitized cisplatin-resistant cells to the cisplatin cytotoxic effect. TFP (10-45 µM) alone elicited dose-dependent cytotoxicity, apoptosis, and G0/G1 arrest on T24/R cells. Co-treatment of TFP potentiated cisplatin-induced cytotoxicity in T24/R cells. The phenomenon that TFP alleviated cisplatin resistance to T24/R was accompanied with concurrent suppression of Bcl-xL. In vivo models confirmed that TFP alone effectively suppressed the T24/R xenograft in nude mice. TFP co-treatment enhanced the antitumor effect of cisplatin on the T24/R xenograft. Our results demonstrated that TFP effectively inhibited cisplatin-resistant UCs and circumvented cisplatin resistance with concurrent Bcl-xL downregulation. These findings provide a promising insight to develop a therapeutic strategy for chemoresistant UCs.


Assuntos
Antipsicóticos/farmacologia , Carcinoma/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Trifluoperazina/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Proteína bcl-X/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antipsicóticos/uso terapêutico , Apoptose , Carcinoma/metabolismo , Linhagem Celular , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Regulação para Baixo , Humanos , Camundongos , Trifluoperazina/uso terapêutico , Neoplasias da Bexiga Urinária/metabolismo , Urotélio/efeitos dos fármacos , Urotélio/metabolismo , Urotélio/patologia , Proteína bcl-X/genética
15.
Cell Oncol (Dordr) ; 42(4): 521-536, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30980364

RESUMO

PURPOSE: Although EGFR activation is a hallmark of glioblastoma (GBM), anti-EGFR therapy has so far not yielded the desired effects. Targeting PI3K/Akt has been proposed as a strategy to increase the cellular sensitivity to EGFR inhibitors. Here we evaluated the contribution of FOXO3a, a key Akt target, in the response of GBM cells to EGFR inhibition. METHODS: FOXO3a activation was assessed by immunofluorescence and gene reporter assays, and by evaluating target gene expression using Western blotting and qRT-PCR. Cellular effects were evaluated using cell viability and apoptosis assays, i.e., Annexin V/PI staining and caspase 3/7 activity measurements. Drug synergism was evaluated by performing isobolographic analyses. Gene silencing experiments were performed using stable shRNA transfections. RESULTS: We found that EGFR inhibition in GBM cells led to FOXO3a activation and to transcriptional modulation of its key targets, including repression of the oncogene FOXM1. In addition, we found that specific FOXO3a activation recapitulated the molecular effects of EGFR inhibition, and that the FOXO3a activator trifluoperazine, a FDA-approved antipsychotic agent, reduced GBM cell growth. Subsequent isobolographic analyses of combination experiments indicated that trifluoperazine and erlotinib cooperated synergistically and that their concomitant treatment induced a robust activation of FOXO3a, leading to apoptosis in GBM cells. Using gene silencing, we found that FOXO3a is essential for the response of GBM cells to EGFR inhibition. CONCLUSIONS: Our data indicate that FOXO3a activation is a crucial event in the response of GBM cells to EGFR inhibition, suggesting that FOXO3a may serve as an actionable therapeutic target that can be modulated using FDA-approved drugs.


Assuntos
Neoplasias Encefálicas/metabolismo , Receptores ErbB/antagonistas & inibidores , Proteína Forkhead Box O3/metabolismo , Glioblastoma/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/metabolismo , Cloridrato de Erlotinib/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Ativação Transcricional/genética , Trifluoperazina/farmacologia , Trifluoperazina/uso terapêutico
16.
J Mol Med (Berl) ; 97(5): 647-658, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30848296

RESUMO

Sepsis is a disease responsible for the death of almost all critical patients. Once infected by virus or bacteria, patients can die due to systemic inflammation within a short period of time. Cytokine storm plays an essential role in causing organ dysfunction and septic shock. Thus, inhibition of cytokine secretion is considered very important in sepsis therapy. In this study, we found that TFP, an antipsychotic drug mainly used to treat schizophrenia by suppressing dopamine secretion, inhibited cytokine release from activated immune cells both in vitro and in vivo. Trifluoperazine (TFP) decreased the levels of pro-inflammatory cytokines without altering their transcription level. In LPS-induced endotoxemia and cecal content injection (CCI) models, TFP intraperitoneal administration improved survival rate. Thus, TFP was considered to inhibit the secretion of proteins through a mechanism similar to that of W7, a calmodulin inhibitor. Finally, we confirmed that TFP treatment relieved organ damage by estimating the concentrations of aspartate transaminase (AST), alanine transaminase (ALT), and blood urea nitrogen (BUN) in the serum. Our findings were regarded as a new discovery of the function of TFP in treating sepsis patients. KEY MESSAGES: • TFP inhibits LPS-induced activation of DCs by suppressing pro-inflammatory cytokine. • Treatment of TFP increases survival of LPS-induced endotoxemia and CCI sepsis models. • TFP exerted a protective effect against tissue or organ damage in animal models.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antipsicóticos/uso terapêutico , Reposicionamento de Medicamentos , Sepse/tratamento farmacológico , Trifluoperazina/uso terapêutico , Animais , Células Cultivadas , Citocinas/análise , Citocinas/imunologia , Feminino , Lipopolissacarídeos/imunologia , Camundongos Endogâmicos C57BL , Sepse/imunologia , Sepse/patologia
17.
Int J Mol Med ; 42(6): 3300-3308, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30272254

RESUMO

As a tumor suppressor, Forkhead box O1 (FOXO1) is located in the nucleus where it regulates gene expression and inhibits tumor progression. However, the antitumor effects of FOXO1 are attenuated in several tumors due to its translocation from the nucleus to the cytoplasm. Trifluoperazine (TFP) is able to reverse tumor drug resistance by inhibiting multidrug resistance (MDR), however, the detailed molecular mechanisms by which this occurs remain to be fully elucidated. In the present study, the doxorubicin (DOX)­resistant SHG44/DOX glioma cell line was established. The results showed that TFP promoted DOX­induced cytotoxicity, cell cycle arrest and early apoptosis using a Cell Counting Kit­8 and flow cytometry. In vivo experiments also demonstrated that DOX combined with TFP reduced tumor volumes and proliferation indices, and led to higher protein levels of FOXO1. In addition, TFP inhibited the nuclear exclusion of FOXO1, contributing toward the downregulation of MDR genes and an increase in intracellular DOX concentrations by reverse transcription­quantitative polymerase chain reaction, western blot analysis, immunofluorescence and spectrophotometer analysis. Therefore, TFP may inhibit DOX resistance by stimulating FOXO1 nuclear translocation and suppressing MDF genes in SHG44/DOX cells, contributing to promising clinical prospects for tumor chemotherapy.


Assuntos
Proteína Forkhead Box O1/metabolismo , Glioma/tratamento farmacológico , Glioma/metabolismo , Trifluoperazina/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Citometria de Fluxo , Proteína Forkhead Box O1/genética , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus
18.
Biomol Concepts ; 9(1): 53-63, 2018 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-29779013

RESUMO

Treatment with analogues of the SERCA-inhibitor Thapsigargin is a promising new approach for a wide variety of cancer entities. However, our previous studies on various tumor cells suggested resistance of SEC62 over-expressing tumors to this treatment. Therefore, we proposed the novel concept that e.g. lung-, prostate-, and thyroid-cancer patients should be tested for SEC62 over-expression, and developed a novel therapeutic strategy for a combinatorial treatment of SEC62 over-expressing tumors. The latter was based on the observations that treatment of SEC62 over-expressing tumor cells with SEC62-targeting siRNAs showed less resistance to Thapsigargin as well as a reduction in migratory potential and that the siRNA effects can be mimicked by the Calmodulin antagonist Trifluoperazine. Therefore, the combinatorial treatment of SEC62 over-expressing tumors was proposed to involve Thapsigargin and Trifluoperazine. Here, we addressed the impact of Thapsigargin and Trifluoperazine in separate and combined treatments of heterotopic tumors, induced by inoculation of human hypopharyngeal squamous cell carcinoma (FaDu)-cells into the mouse flank. Seeding of the tumor cells and/or their growth rate were significantly reduced by all three treatments, suggesting Trifluoperazine is a small molecule to be considered for future therapeutic strategies for patients, suffering from Sec62-overproducing tumors.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias Hipofaríngeas/tratamento farmacológico , Proteínas de Membrana Transportadoras/metabolismo , Tapsigargina/uso terapêutico , Trifluoperazina/uso terapêutico , Animais , Calmodulina/antagonistas & inibidores , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Proliferação de Células , Inibidores Enzimáticos/sangue , Neoplasias de Cabeça e Pescoço/genética , Humanos , Neoplasias Hipofaríngeas/genética , Camundongos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , Carcinoma de Células Escamosas de Cabeça e Pescoço , Tapsigargina/sangue , Trifluoperazina/sangue
19.
Eur J Med Chem ; 151: 186-198, 2018 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-29614416

RESUMO

Repositioning of the antipsychotic drug trifluoperazine for treatment of glioblastoma, an aggressive brain tumor, has been previously suggested. However, trifluoperazine did not increase the survival time in mice models of glioblastoma. In attempt to identify an effective trifluoperazine analog, fourteen compounds have been synthesized and biologically in vitro and in vivo assessed. Using MTT assay, compounds 3dc and 3dd elicited 4-5 times more potent inhibitory activity than trifluoperazine with IC50 = 2.3 and 2.2 µM against U87MG glioblastoma cells, as well as, IC50 = 2.2 and 2.1 µM against GBL28 human glioblastoma patient derived primary cells, respectively. Furthermore, they have shown a reasonable selectivity for glioblastoma cells over NSC normal neural cell. In vivo evaluation of analog 3dc confirmed its advantageous effect on reduction of tumor size and increasing the survival time in brain xenograft mouse model of glioblastoma. Molecular modeling simulation provided a reasonable explanation for the observed variation in the capability of the synthesized analogs to increase the intracellular Ca2+ levels. In summary, this study presents compound 3dc as a proposed new tool for the adjuvant chemotherapy of glioblastoma.


Assuntos
Antineoplásicos/uso terapêutico , Antipsicóticos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Trifluoperazina/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Antipsicóticos/química , Antipsicóticos/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Reposicionamento de Medicamentos , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Camundongos , Simulação de Acoplamento Molecular , Trifluoperazina/análogos & derivados , Trifluoperazina/farmacologia , Células Tumorais Cultivadas
20.
Int J Mol Med ; 41(6): 3278-3286, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29568865

RESUMO

The inhibition of mesangial cell (MC) proliferation has become an important therapy in preventing glomerular proliferation diseases. Trifluoperazine (TFP) has been reported to inhibit the proliferation of several types of cancer cell, however, the effects of TFP in renal proliferation diseases remain to be fully elucidated. The present study examined the effects of TFP on the proliferation of MCs and quantified cell apoptosis progression in vivo and in vitro. The effects of various TFP concentrations and treatment durations on cell proliferation and cell apoptosis in vitro were analyzed using flow cytometry in conjunction with a Cell Counting kit­8 assay. Cell proliferation in vivo was determined using hematoxylin and eosin staining and immunohistochemistry of Ki67. The expression of the two cell apoptosis­related proteins, B­cell lymphoma-2 (Bcl­2) and Bcl­2­associated X protein (Bax), were estimated using western blot analysis and immunohistochemistry in vivo and in vitro. TFP­induced phosphatidylinositol 3­kinase (PI3K)/protein kinase B (AKT) signaling pathways were also estimated using western blot analysis. These results suggested that TFP inhibited MC proliferation in a dose­ and time­dependent manner. It was found that TFP inhibited the abnormal proliferation of MCs, which was stimulated by 20% fetal bovine serum in vitro and in lupus MRL/lpr mice. TFP promoted cell apoptosis, downregulated the expression of Bcl­2 and upregulated the expression of Bax in a dose­dependent manner at mRNA and protein levels. In addition, TFP inhibited phosphorylated AKT, potentially leading to the suppressed activation of PI3K/AKT signaling pathways. TFP treatment significantly decreased the levels of blood urea nitrogen and serum creatinine, but had no significant effects on the body weight and liver function of the lupus mice. These results validated and reinforced the potential of TFP in the treatment of mesangial proliferative diseases.


Assuntos
Nefrite Lúpica/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Trifluoperazina/uso terapêutico , Proteína X Associada a bcl-2/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Nefrite Lúpica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...