Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Ovarian Res ; 17(1): 71, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38566237

RESUMO

BACKGROUND: Ovarian cancer remains the deadliest of the gynecologic cancers in the United States. There have been limited advances in treatment strategies that have seen marked increases in overall survival. Thus, it is essential to continue developing and validating new treatment strategies and markers to identify patients who would benefit from the new strategy. In this report, we sought to further validate applications for a novel humanized anti-Sialyl Tn antibody-drug conjugate (anti-STn-ADC) in ovarian cancer. METHODS: We aimed to further test a humanized anti-STn-ADC in sialyl-Tn (STn) positive and negative ovarian cancer cell line, patient-derived organoid (PDO), and patient-derived xenograft (PDX) models. Furthermore, we sought to determine whether serum STn levels would reflect STn positivity in the tumor samples enabling us to identify patients that an anti-STn-ADC strategy would best serve. We developed a custom ELISA with high specificity and sensitivity, that was used to assess whether circulating STn levels would correlate with stage, progression-free survival, overall survival, and its value in augmenting CA-125 as a diagnostic. Lastly, we assessed whether the serum levels reflected what was observed via immunohistochemical analysis in a subset of tumor samples. RESULTS: Our in vitro experiments further define the specificity of the anti-STn-ADC. The ovarian cancer PDO, and PDX models provide additional support for an anti-STn-ADC-based strategy for targeting ovarian cancer. The custom serum ELISA was informative in potential triaging of patients with elevated levels of STn. However, it was not sensitive enough to add value to existing CA-125 levels for a diagnostic. While the ELISA identified non-serous ovarian tumors with low CA-125 levels, the sample numbers were too small to provide any confidence the STn ELISA would meaningfully add to CA-125 for diagnosis. CONCLUSIONS: Our preclinical data support the concept that an anti-STn-ADC may be a viable option for treating patients with elevated STn levels. Moreover, our STn-based ELISA could complement IHC in identifying patients with whom an anti-STn-based strategy might be more effective.


Assuntos
Neoplasias dos Genitais Femininos , Neoplasias Ovarianas , Humanos , Feminino , Antígenos Glicosídicos Associados a Tumores/metabolismo , Antígeno Ca-125 , Ensaio de Imunoadsorção Enzimática , Biomarcadores Tumorais
2.
Br J Cancer ; 130(9): 1463-1476, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38438589

RESUMO

BACKGROUND: Uterine serous cancer (USC) comprises around 10% of all uterine cancers. However, USC accounts for approximately 40% of uterine cancer deaths, which is attributed to tumor aggressiveness and limited effective treatment. Galectin 3 (Gal3) has been implicated in promoting aggressive features in some malignancies. However, Gal3's role in promoting USC pathology is lacking. METHODS: We explored the relationship between LGALS3 levels and prognosis in USC patients using TCGA database, and examined the association between Gal3 levels in primary USC tumors and clinical-pathological features. CRISPR/Cas9-mediated Gal3-knockout (KO) and GB1107, inhibitor of Gal3, were employed to evaluate Gal3's impact on cell function. RESULTS: TCGA analysis revealed a worse prognosis for USC patients with high LGALS3. Patients with no-to-low Gal3 expression in primary tumors exhibited reduced clinical-pathological tumor progression. Gal3-KO and GB1107 reduced cell proliferation, stemness, adhesion, migration, and or invasion properties of USC lines. Furthermore, Gal3-positive conditioned media (CM) stimulated vascular tubal formation and branching and transition of fibroblast to cancer-associated fibroblast compared to Gal3-negative CM. Xenograft models emphasized the significance of Gal3 loss with fewer and smaller tumors compared to controls. Moreover, GB1107 impeded the growth of USC patient-derived organoids. CONCLUSION: These findings suggest inhibiting Gal3 may benefit USC patients.


Assuntos
Proteínas Sanguíneas , Cistadenocarcinoma Seroso , Galectina 3 , Neoplasias Uterinas , Humanos , Feminino , Neoplasias Uterinas/patologia , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Galectina 3/genética , Galectina 3/metabolismo , Cistadenocarcinoma Seroso/patologia , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Proliferação de Células , Linhagem Celular Tumoral , Prognóstico , Animais , Camundongos , Galectinas/genética , Galectinas/metabolismo , Movimento Celular
3.
Cell ; 186(11): 2361-2379.e25, 2023 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-37192619

RESUMO

Multiple anticancer drugs have been proposed to cause cell death, in part, by increasing the steady-state levels of cellular reactive oxygen species (ROS). However, for most of these drugs, exactly how the resultant ROS function and are sensed is poorly understood. It remains unclear which proteins the ROS modify and their roles in drug sensitivity/resistance. To answer these questions, we examined 11 anticancer drugs with an integrated proteogenomic approach identifying not only many unique targets but also shared ones-including ribosomal components, suggesting common mechanisms by which drugs regulate translation. We focus on CHK1 that we find is a nuclear H2O2 sensor that launches a cellular program to dampen ROS. CHK1 phosphorylates the mitochondrial DNA-binding protein SSBP1 to prevent its mitochondrial localization, which in turn decreases nuclear H2O2. Our results reveal a druggable nucleus-to-mitochondria ROS-sensing pathway-required to resolve nuclear H2O2 accumulation and mediate resistance to platinum-based agents in ovarian cancers.


Assuntos
Antineoplásicos , Espécies Reativas de Oxigênio , Antineoplásicos/farmacologia , Antineoplásicos/metabolismo , Peróxido de Hidrogênio/metabolismo , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Núcleo Celular/metabolismo , Humanos
4.
Toxicol In Vitro ; 89: 105579, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36870549

RESUMO

Malignant melanoma is considered a deadly aggressive form of skin cancer that frequently metastasizes to various distal organs, which harbors mutations of the BRAF or NRAS which occur in 30 to 50% of melanoma patients. The growth factors secreted by melanoma cells contribute to tumor angiogenesis with the acquisition of metastatic potential by epithelial-mesenchymal transition (EMT) and drive melanoma growth toward a more aggressive form. Niclosamide (NCL) is an FDA-approved anthelmintic drug and is reported to have strong anti-cancer properties against various solid and liquid tumors. Its role in BRAF or NRAS mutated cells is unknown. In this context, we uncovered the role of NCL in impeding malignant metastatic melanoma in vitro in SK-MEL-2 and SK-MEL-28 cell lines. We found that NCL induces significant ROS generation and apoptosis through a series of molecular mechanisms, such as depolarization of mitochondrial membrane potential, arresting the cell cycle at the sub G1 phase with a significant increase in the DNA cleavage via topoisomerase II in both cell lines. We also found that NCL potently inhibited metastasis, which was examined by scratch wound assay, Additionally, we found that NCL inhibits the most important markers involved in the EMT signaling cascade that are stimulated by TGF-ß such as N-cadherin, Snail, Slug, Vimentin, α-SMA and p-Smad 2/3. This work provides useful insights into the mechanism of NCL in BRAF/NRAF mutant melanoma cells via inhibition of molecular signaling events involved in EMT signaling, and apoptosis induction.


Assuntos
Melanoma , Proteínas Proto-Oncogênicas B-raf , Humanos , Proteínas Proto-Oncogênicas B-raf/genética , Niclosamida/farmacologia , Transição Epitelial-Mesenquimal , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Apoptose , Linhagem Celular Tumoral , Mutação , Proteínas de Membrana/genética , GTP Fosfo-Hidrolases/genética
5.
bioRxiv ; 2023 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-36945474

RESUMO

Multiple chemotherapies are proposed to cause cell death in part by increasing the steady-state levels of cellular reactive oxygen species (ROS). However, for most of these drugs exactly how the resultant ROS function and are sensed is poorly understood. In particular, it's unclear which proteins the ROS modify and their roles in chemotherapy sensitivity/resistance. To answer these questions, we examined 11 chemotherapies with an integrated proteogenomic approach identifying many unique targets for these drugs but also shared ones including ribosomal components, suggesting one mechanism by which chemotherapies regulate translation. We focus on CHK1 which we find is a nuclear H 2 O 2 sensor that promotes an anti-ROS cellular program. CHK1 acts by phosphorylating the mitochondrial-DNA binding protein SSBP1, preventing its mitochondrial localization, which in turn decreases nuclear H 2 O 2 . Our results reveal a druggable nucleus-to-mitochondria ROS sensing pathway required to resolve nuclear H 2 O 2 accumulation, which mediates resistance to platinum-based chemotherapies in ovarian cancers.

6.
Int Immunopharmacol ; 115: 109701, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36641892

RESUMO

Acute respiratory distress syndrome (ARDS) is associated with severe lung inflammation, edema, hypoxia, and high vascular permeability. The COVID-19-associated pandemic ARDS caused by SARS-CoV-2 has created dire global conditions and has been highly contagious. Chronic inflammatory disease enhances cancer cell proliferation, progression, and invasion. We investigated how acute lung inflammation activates the tumor microenvironment and enhances lung metastasis in LPS induced in vitro and in vivo models. Respiratory illness is mainly caused by cytokine storm, which further influences oxidative and nitrosative stress. The LPS-induced inflammatory cytokines made the conditions suitable for the tumor microenvironment in the lungs. In the present study, we observed that LPS induced the cytokine storm and promoted lung inflammation via BRD4, which further caused the nuclear translocation of p65 NF-κB and STAT3. The transcriptional activation additionally triggers the tumor microenvironment and lung metastasis. Thus, BRD4-regulated p65 and STAT3 transcriptional activity in ARDS enhances lung tumor metastasis. Moreover, LPS-induced ARDS might promote the tumor microenvironment and increase cancer metastasis into the lungs. Collectively, BRD4 plays a vital role in inflammation-mediated tumor metastasis and is found to be a diagnostic and molecular target in inflammation-associated cancers.


Assuntos
COVID-19 , Neoplasias Pulmonares , Pneumonia , Síndrome do Desconforto Respiratório , Humanos , Proteínas Nucleares/genética , Lipopolissacarídeos/farmacologia , Microambiente Tumoral , Síndrome da Liberação de Citocina , SARS-CoV-2 , Fatores de Transcrição/genética , Pulmão/patologia , Síndrome do Desconforto Respiratório/induzido quimicamente , Pneumonia/induzido quimicamente , Inflamação , Proteínas de Ciclo Celular/genética
7.
J Nanobiotechnology ; 19(1): 101, 2021 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-33849555

RESUMO

BACKGROUND: Psoriasis is a chronic autoimmune skin disease characterized by hyperproliferation of keratinocytes. Wide treatment options used to treat psoriasis is associated with various adverse effects. To overcome this nanoformulation is prepared. Selenium is an essential trace element and plays major role in oxidation reduction system. Toxicity and stability limits the applications of selenium. Toxicity can be reduced and stabilized upon preparation into nanoparticles. RESULTS: Selenium nanoparticles (SeNPs) exhibit potent apoptosis through the generation of reactive oxygen species (ROS) with cell cycle arrest. SeNPs topical gel application produced significant attenuation of psoriatic severity with the abrogation of acanthosis and splenomegaly. SeNPs reduced the phosphorylation and expressions of MAPKs, STAT3, GSK-3ß, Akt along with PCNA, Ki67, and cyclin-D1. CONCLUSION: SeNPs inhibit various inflammation and proliferation mediated pathways and could be an ideal candidate for psoriasis therapy. MATERIALS AND METHODS: SeNPs were characterized and various techniques were used to determine apoptosis and other molecular mechanisms. In vivo studies were performed by inducing psoriasis with imiquimod (IMQ). SeNPs were administered via topical route.


Assuntos
Inflamação/tratamento farmacológico , Nanopartículas/química , Psoríase/tratamento farmacológico , Selênio/química , Selênio/farmacologia , Animais , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Imiquimode , Queratinócitos , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/uso terapêutico , Oxirredução , Fosforilação , Espécies Reativas de Oxigênio , Fator de Transcrição STAT3/metabolismo
8.
Int J Pharm ; 601: 120536, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33781885

RESUMO

Acute respiratory distress syndrome (ARDS) is a life threatening respiratory disease associated with pulmonary edema, alveolar dysfunction, hypoxia, and inflammatory cell accumulation. The most contagious form of COVID-19 associated with ARDS caused by SARS-CoV-2. SARS-CoV-2 majorly produces the cytokine storm and severe lung inflammation and ultimately leads to respiratory failure. ARDS is a complex disease and there is no proper therapeutics for effective therapy. Still, there is a huge scope to identify novel targets to combat respiratory illness. In the current study, we have identified the epigenetic regulating protein BRD4 and developed siRNA based nanomedicine to treat the ARDS. The liposomes were prepared by thin-film hydration method, where BRD4 siRNA complexed with cationic lipid and exhibited 96.24 ± 18.01 nm size and stable even in the presence of RNase. BRD4 siRNA lipoplexes (BRD4-siRNA-LP) inhibited inflammatory cells in lungs and suppressed the lipopolysaccharide (LPS) induced the neutrophil infiltration and mast cell accumulation. Also, BRD4 siRNA based nanomedicine significantly reduced the LPS induced cytokine storm followed by inflammatory signaling pathways. Interestingly, BRD4-siRNA-LP suppressed the LPS-induced p65 and STAT3 nuclear translocation and ameliorated the lung inflammation. Thus, BRD4-siRNA-LP could be a plausible therapeutic option for treating ARDS and might be useful for combating the COVID-19 associated respiratory illness.


Assuntos
COVID-19 , Síndrome do Desconforto Respiratório , Proteínas de Ciclo Celular , Humanos , Lipopolissacarídeos , Proteínas Nucleares , Síndrome do Desconforto Respiratório/tratamento farmacológico , SARS-CoV-2 , Fatores de Transcrição/genética
9.
Nanomedicine ; 33: 102351, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33418136

RESUMO

Acute respiratory distress syndrome (ARDS) is a deadly respiratory illness associated with refractory hypoxemia and pulmonary edema. The recent pandemic outbreak of COVID-19 is associated with severe pneumonia and inflammatory cytokine storm in the lungs. The anti-inflammatory phytomedicine nimbolide (NIM) may not be feasible for clinical translation due to poor pharmacokinetic properties and lack of suitable delivery systems. To overcome these barriers, we have developed nimbolide liposomes conjugated with iRGD peptide (iRGD-NIMLip) for targeting lung inflammation. It was observed that iRGD-NIMLip treatment significantly inhibited oxidative stress and cytokine storm compared to nimbolide free-drug (f-NIM), nimbolide liposomes (NIMLip), and exhibited superior activity compared to dexamethasone (DEX). iRGD-NIMLip abrogated the LPS induced p65 NF-κB, Akt, MAPK, Integrin ß3 and ß5, STAT3, and DNMT1 expression. Collectively, our results demonstrate that iRGD-NIMLip could be a promising novel drug delivery system to target severe pathological consequences observed in ARDS and COVID-19 associated cytokine storm.


Assuntos
Anti-Inflamatórios/administração & dosagem , Limoninas/administração & dosagem , Lipossomos/química , Oligopeptídeos/química , Síndrome do Desconforto Respiratório/tratamento farmacológico , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/uso terapêutico , Linhagem Celular , Sistemas de Liberação de Medicamentos , Endotoxinas , Humanos , Limoninas/química , Limoninas/uso terapêutico , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Síndrome do Desconforto Respiratório/induzido quimicamente , Síndrome do Desconforto Respiratório/patologia
10.
Bioorg Chem ; 102: 104125, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32738568

RESUMO

In this study, we have synthesized a new series of benzimidazole-triazole hybrids as galectin-1 (gal-1) mediated apoptosis-inducing agents, and evaluated for their potential anticancer activity against a panel of human cancer cell lines viz. breast cancer (MCF-7 and MDA-MB-231) lung cancer (A-549 and NCI-H460), and human keratinocyte cancer (HaCaT), using MTT assay. The target compound 7c exhibited an excellent growth inhibition against lung cancer (A-549 and NCI-H460) cells with an IC50 value of 0.63 ± 0.21 µM, and 0.99 ± 0.01 µM respectively. The target compound 7c also showed a significant growth inhibition against breast cancer (MCF-7 and MDA-MB-23) with an IC50 value of 1.3 ± 0.18 µM, and 0.94 ± 0.02 µM respectively. In addition, the radiochemical synthesis has been performed using fluorine-18 radionuclide in the GE Tracer-lab FX2N module to prove the target compound 7c as a PET imaging agent. In the final stage, the 18F-7c target compound was successfully purified with 60% ethanol in water. The radiochemical purity was achieved >95% using HPLC, and the residual solvent DMF limit was around 78 ± 3 ppm confirmed by GC analysis. Further, the apoptosis induction by 7c in lung cancer (A-549) cells was confirmed as a result of the decrease in MMP levels, increased percentage of apoptotic cells, and sub G1 phase arrest by JC-1 staining, DAPI staining, annexin V-FITC/PI, and flow cytometric analysis. In addition, the target compound 7c significantly reduced the gal-1 protein levels in a dose-dependent manner as confirmed by ELISA studies. The protein binding studies like Surface Plasmon Resonance (SPR) and Fluorescence Spectroscopy (FS) studies indicated that the target compound 7c is capable of binding to gal-1 with an equilibrium constant (KD) value of 1.19E-06 M, and binding constant (Ka) of 9.5 × 103 M-1 respectively. The in-silico computational studies also revealed possible interactions and pharmacokinetic properties (ADMET) of compound 7c with the binding domain of gal-1. Therefore, the novel benzimidazole-triazole hybrids as apoptosis-inducing agents in lung cancer would be potential cytotoxic and PET imaging agents via gal-1.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzimidazóis/farmacologia , Desenho de Fármacos , Galectina 1/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Triazóis/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Benzimidazóis/química , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Radioisótopos de Flúor , Galectina 1/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Estrutura Molecular , Relação Estrutura-Atividade , Triazóis/química , Células Tumorais Cultivadas
11.
Pharm Dev Technol ; 25(8): 971-988, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32403972

RESUMO

Clinical significance of Rutin (RUT) is limited by poor dissolution rate and low oral bioavailability. The study was designed to improve the physicochemical and therapeutic potential of the drug by formulating nanosuspension (NS) for osteoporosis. Rutin nanosuspension (RUT-NS) was prepared after screening a range of stabilizers and their combinations at a different concentration by antisolvent precipitation technique. Effect of precipitation on crystallinity (differential scanning calorimetry DSC, X-ray diffraction studies XRD), morphology (scanning electron microscopy, SEM) and chemical interaction (attenuated total reflectance fourier-transform infrared spectroscopy ATR-FTIR) were studied through biophysical techniques. An optimized nanosuspension exhibited a minimum particle size of 122.85 ± 5.02 nm with higher dissolution of RUT-NS (87. 63 ± 2.29%) as compared to pure drug (39.77 ± 2.8 6%). The enhanced intestine absorption and apparent permeability were achieved due to the improved particle size, surface area and dissolution. RUT-NS displayed greater (3 folds) AUC0-24 h than pure drug. In vitro assays with RUT-NS depicted an increased cell proliferation, antioxidant (ROS) activity and osteocalcin production in MG-63 osteoblast cells. The augmented biochemical in vivo biomarkers and bone quality proved the protective effect of RUT-NS. The results supported RUT-NS as a potential therapy for maintaining bone health.


Assuntos
Nanopartículas/administração & dosagem , Osteoporose/tratamento farmacológico , Osteoporose/metabolismo , Rutina/metabolismo , Rutina/farmacologia , Suspensões/metabolismo , Suspensões/farmacologia , Administração Oral , Animais , Disponibilidade Biológica , Varredura Diferencial de Calorimetria/métodos , Química Farmacêutica/métodos , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos/fisiologia , Excipientes/química , Feminino , Absorção Intestinal/fisiologia , Microscopia Eletrônica de Varredura , Nanopartículas/metabolismo , Tamanho da Partícula , Permeabilidade , Ratos , Ratos Wistar , Solubilidade/efeitos dos fármacos , Difração de Raios X/métodos
12.
Life Sci ; 254: 117765, 2020 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32437797

RESUMO

The ongoing wreaking global outbreak of the novel human beta coronavirus (CoV) pathogen was presumed to be from a seafood wholesale market in Wuhan, China, belongs to the Coronaviridae family in the Nidovirales order. The virus is highly contagious with potential human-human transmission which was named as the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has spread across six continents and emerged as a global pandemic in short span with alarming levels of spread and severity. This virus associated symptoms and infectious respiratory illness is designated as coronavirus disease 19 (COVID-19). The SARS-CoV-2 possesses enveloped club-like spike protein projections with positive-sense large RNA genome and has a unique replication strategy. This virus was believed to have zoonotic origin with genetical identity to bat and pangolin CoV. In the current review, we introduce a general overview about the human CoVs and the associated diseases, the origin, structure, replication and key clinical events that occur in the COVID-19 pathogenicity. Furthermore, we focused on possible therapeutic options such as repurposing drugs including antimalarials, antivirals, antiparasitic drugs, and anti-HIV drugs, as well as monoclonal antibodies, vaccines as potential treatment options. Also we have summarized the latest research progress on the usage of stem cell therapy, human convalescent serum, interferon's, in the treatment of COVID-19.


Assuntos
Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/terapia , Pneumonia Viral/epidemiologia , Pneumonia Viral/terapia , Animais , Betacoronavirus/isolamento & purificação , COVID-19 , Infecções por Coronavirus/virologia , Pandemias/prevenção & controle , Pneumonia Viral/virologia , SARS-CoV-2 , Zoonoses/epidemiologia , Zoonoses/terapia , Zoonoses/virologia
13.
Cell Death Dis ; 11(1): 21, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31924750

RESUMO

Psoriasis is an autoimmune skin disease, where chronic immune responses due to exaggerated cytokine signaling, abnormal differentiation, and evasion of keratinocytes apoptosis plays a crucial role in mediating abnormal keratinocytes hyperproliferation. From the therapeutic perspective, the molecules with strong anti-proliferative and anti-inflammatory properties could have tremendous relevance. In this study, we demonstrated that piperlongumine (PPL) treatment effectively abrogated the hyperproliferation and differentiation of keratinocytes by inducing ROS-mediated late apoptosis with loss of mitochondrial membrane potential. Besides, the arrest of cell cycle was found at Sub-G1 phase as a result of DNA fragmentation. Molecularly, inhibition of STAT3 and Akt signaling was observed with a decrease in proliferative markers such as PCNA, ki67, and Cyclin D1 along with anti-apoptotic Bcl-2 protein expression. Keratin 17 is a critical regulator of keratinocyte differentiation, and it was found to be downregulated with PPL significantly. Furthermore, prominent anti-inflammatory effects were observed by inhibition of lipopolysaccharide (LPS)/Imiquimod (IMQ)-induced p65 NF-κB signaling cascade and strongly inhibited the production of cytokine storm involved in psoriasis-like skin inflammation, thus led to the restoration of normal epidermal architecture with reduction of epidermal hyperplasia and splenomegaly. In addition, PPL epigenetically inhibited histone-modifying enzymes, which include histone deacetylases (HDACs) of class I (HDAC1-4) and class II (HDAC6) evaluated by immunoblotting and HDAC enzyme assay kit. In addition, our results show that PPL effectively inhibits the nuclear translocation of p65 and a histone modulator HDAC3, thus sequestered in the cytoplasm of macrophages. Furthermore, PPL effectively enhanced the protein-protein interactions of HDAC3 and p65 with IκBα, which was disrupted by LPS stimulation and were evaluated by Co-IP and molecular modeling. Collectively, our findings indicate that piperlongumine may serve as an anti-proliferative and anti-inflammatory agent and could serve as a potential therapeutic option in treating psoriasis.


Assuntos
Dioxolanos/uso terapêutico , Epigênese Genética , Inflamação/tratamento farmacológico , Psoríase/tratamento farmacológico , Psoríase/genética , Pele/patologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiocinas/metabolismo , Dioxolanos/farmacologia , Epiderme/patologia , Epigênese Genética/efeitos dos fármacos , Células HaCaT , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Imiquimode/efeitos adversos , Inflamação/genética , Queratina-17/metabolismo , Queratinócitos/efeitos dos fármacos , Queratinócitos/patologia , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Psoríase/induzido quimicamente , Psoríase/patologia , Células RAW 264.7 , Fator de Transcrição STAT3/metabolismo
14.
J Cell Physiol ; 235(6): 5270-5283, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31846070

RESUMO

Drug discovery is an onerous, extremely expensive, and time-consuming process. Instead, drug repurposing is an attractive strategy for exploiting novel indications for a drug beyond its original use. The untapped potential of drug repurposing compensates the barriers associated with the drug discovery pipeline. Psoriasis is an autoimmune skin disease, where hyperproliferation of keratinocytes and exaggerated immune responses are the important hallmarks of the disease. Extensive in vitro and preclinical research has demonstrated that niclosamide was found to exert potent anticancer and anti-inflammatory properties by targeting STAT3, p65 NF-κB, and NFATc-1 signaling paradigm with minimal host toxicity. From the disease perspective, the static intracellular molecular network in both cancer and psoriasis share overlapping pathological features in terms of hyperproliferation and chronic inflammation, which is mediated by the aforementioned signaling cascade. The plausible mechanistic relevance has prompted us to investigate the implementation of niclosamide for repositioning in psoriasis. Our in vitro and in vivo findings suggest that niclosamide inhibits keratinocytes hyperproliferation by reactive oxygen species-mediated apoptosis through the loss of mitochondrial membrane potential, cell cycle arrest at Sub G1 phase, and DNA fragmentation. Furthermore, niclosamide treatment resulted in abrogation of lipopolysaccharide-induced inflammatory cytokine levels in murine macrophages. Additionally, our results provided a preclinical rationale in imiquimod (IMQ)-induced BALB/c mouse model, where niclosamide diligently mitigated the IMQ-induced epidermal hyperplasia and inflammation by downregulating STAT3, p65 NF-κB, and NFATc-1 transcription factors along with Akt, Ki-67, and ICAM-1 protein expression.


Assuntos
Inflamação/tratamento farmacológico , Fatores de Transcrição NFATC/genética , Fator de Transcrição STAT3/genética , Dermatopatias/tratamento farmacológico , Fator de Transcrição RelA/genética , Animais , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Reposicionamento de Medicamentos , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Queratinócitos/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Niclosamida/farmacologia , Psoríase/tratamento farmacológico , Psoríase/patologia , Transdução de Sinais/efeitos dos fármacos , Dermatopatias/patologia
15.
Bioorg Chem ; 93: 103306, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31586710

RESUMO

A facile synthetic strategy has been developed for the generation of structurally diverse N-fused heterocycles. The formation of fused 1,2,3-triazole indolo and pyrrolodiazepines proceeds through an initial Knoevenagel condensation followed by intramolecular azide-alkyne cycloaddition reaction at room temperature without recourse to the traditional Cu(I)-catalyzed azide-alkyne cycloadditions. The synthesized compounds were evaluated for their in vitro anti-cancer activity against the NCI 60 cell line panel. Among the tested compounds, 3a and 3h were found to exhibit potent inhibitory activity against many of the cell lines. Cell cycle analysis indicated that the compounds inhibit the cell cycle at sub G1 phase. The DNA- nano drop method, viscosity experiment and docking studies suggested these compounds possess DNA binding affinity.


Assuntos
Antineoplásicos/síntese química , Azepinas/química , DNA/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Azepinas/metabolismo , Azepinas/farmacologia , Sítios de Ligação , Catálise , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cobre/química , Reação de Cicloadição , DNA/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Indóis/química , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Conformação Molecular , Simulação de Acoplamento Molecular , Conformação de Ácido Nucleico , Pirróis/química , Triazóis/química
16.
Bioorg Chem ; 92: 103188, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31450167

RESUMO

A series of certain benzyl/phenethyl thiazolidinone-indole hybrids were synthesized for the study of anti-proliferative activity against A549, NCI-H460 (lung cancer), MDA-MB-231 (breast cancer), HCT-29 and HCT-15 (colon cancer) cell lines by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). We found that compound G37 displayed highest cytotoxicity with IC50 value of 0.92 ±â€¯0.12 µM towards HCT-15 cancer cell line among all the synthesized compounds. Moreover, compound G37 was also tested on normal human lung epithelial cells (L132) and was found to be safe in contrast to HCT-15 cells. The lead compound G37 showed significant G2/M phase arrest in HCT-15 cells. Additionally, compound G37 significantly inhibited tubulin polymerization with IC50 value of 2.92 ±â€¯0.23 µM. Mechanistic studies such as acridine orange/ethidium bromide (AO/EB) dual staining, DAPI nuclear staining, annexinV/propidium iodide dual staining, clonogenic growth inhibition assays inferred that compound G37 induced apoptotic cell death in HCT-15 cells. Moreover, loss of mitochondrial membrane potential with elevated intracellular ROS levels was observed by compound G37. These compounds bind at the active pocket of the α/ß-tubulin with higher number of stable hydrogen bonds, hydrophobic and arene-cation interactions confirmed by molecular modeling studies.


Assuntos
Antineoplásicos/síntese química , Benzeno/química , Indóis/síntese química , Tiazolidinas/química , Moduladores de Tubulina/síntese química , Tubulina (Proteína)/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Indóis/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Simulação de Acoplamento Molecular , Estrutura Molecular , Ligação Proteica , Eletricidade Estática , Relação Estrutura-Atividade , Moduladores de Tubulina/farmacologia
17.
Nanomedicine (Lond) ; 14(15): 1991-2010, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31355710

RESUMO

Aim: Selenium nanoparticles (SeNPs) may have a potential role in treating dermal disorders due to its wide therapeutic properties, but there is a need to evaluate its toxicity in keratinocytes. The present study evaluated the molecular mechanism and mode of cell death induced by SeNPs on dermal keratinocytes. Materials & methods: SeNPs were synthesized, characterized and studied in human keratinocytes cells. Oxidative stress and mitochondrial membrane depolarization were evaluated by various techniques. Additionally, autophagy mediated apoptotic cell death was evaluated. Results: SeNPs induced oxidative stress and apoptotic cell death in keratinocytes by increasing autophagy through the formation of acidic lysosomes and autophagosomes. Conclusion: Overall, SeNPs induce the oxidative stress and autophagy mediated apoptotic cell death in human keratinocytes cells.


Assuntos
Morte Celular Autofágica/efeitos dos fármacos , Queratinócitos/efeitos dos fármacos , Nanopartículas , Selênio/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Nanopartículas/química , Nanopartículas/ultraestrutura , Estresse Oxidativo/efeitos dos fármacos , Selênio/química
18.
Chem Biol Drug Des ; 94(5): 1919-1929, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31169963

RESUMO

A series of novel morpholines linked coumarin-triazole hybrids (6a-6v) has been synthesized and evaluated for their anti-proliferative potential on a panel of five human cancer cell lines, namely bone (MG-63), lung (A549), breast (MDA-MB-231), colon (HCT-15) and liver (HepG2), using MTT assay. Among all, the compound 6n {7-((1-(2,4-dichlorobenzyl)-1H-1,2,3-triazol-4-yl) methoxy)-4-((2,6-dimethylmorpholino) methyl)-2H-chromen-2-one} showed significant growth inhibition against MG-63 cells with an IC50 value of 0.80 ± 0.22 µM. Further, induction of apoptosis by 6n of MG-63 cells confirmed as a result of morphological changes, the sub-G1 phase arrest, increased percentage of apoptotic cells, and decrease in mitochondrial membrane potential and increase in reactive oxygen species levels. The in vitro Gal-1 expression in cell culture supernatant of MG-63 cells treated with compound 6n showed dose-dependent reduction. The binding constant (Ka ) of 6n with Gal-1 was calculated from the intercept value which was observed as 3.0 × 105  M-1 by fluorescence spectroscopy. Surface plasmon resonance showed that 6n binds to Gal-1 with binding constant (Ka ) of 1.29E+04 1/Ms and equilibrium constant KD value of 7.54E-07 M, respectively. Molecular docking studies revealed the binding interactions of 6n with Gal-1.


Assuntos
Antineoplásicos/síntese química , Cumarínicos/química , Morfolinas/síntese química , Triazóis/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Galectina 1/química , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Simulação de Acoplamento Molecular , Estrutura Molecular , Morfolinas/farmacologia , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade
19.
Int Immunopharmacol ; 71: 301-312, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30933843

RESUMO

The purpose of this study was to investigate the protective effects of ambroxol in psoriasis-like skin inflammation both in vitro and in vivo and delineate the molecular mechanism of ambroxol. Our data demonstrated that ambroxol has an imperative role in inhibiting the lipopolysaccharide (LPS) stimulated nitrite levels, total cellular and mitochondrial reactive oxygen species level which was determined by Griess assay, DCFDA, and MitoSOX Red staining, respectively. We found that ambroxol remarkably reduced imiquimod (IMQ) induced epidermal hyperplasia, psoriasis area and severity index (PASI) scoring, splenomegaly, skin, and ear fold thickness. In addition, the histopathological evaluation revealed that ambroxol topical and subcutaneous treatment eloquently reduced psoriasiform lesions including acanthosis. Moreover, with ambroxol intervention, the levels of antioxidants glutathione (GSH), superoxide dismutase (SOD), and IL-10 were found to be increased along with a reduction in nitrite levels in skin tissues. On the other hand, ambroxol treatment significantly reduced imiquimod-induced levels of inflammatory cytokines such as IL-1ß, IL-6, IL-17, IL-22, IL-23, TGF-ß, and TNF-α. Furthermore, from immunoblotting, we found a decrease in the protein expression of nitrotyrosine, iNOS, NF-κB and MAPKs signaling cascade with a concomitant increase in the expression of Nrf-2 and SOD-1 in RAW 264.7 cells and skin tissues by ambroxol. Similar findings were observed by immunofluorescence in macrophages. Moreover, ambroxol downregulated the ICAM-1 and Ki67 expression observed in skin tissues. Collectively, our results demonstrate that ambroxol may have intriguing therapeutic possibilities in attenuating psoriasis.


Assuntos
Ambroxol/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Inflamação/tratamento farmacológico , Macrófagos/imunologia , Psoríase/tratamento farmacológico , Pele/patologia , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Hiperplasia , Imiquimode , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Células RAW 264.7 , Transdução de Sinais
20.
Cell Death Dis ; 10(2): 81, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30692512

RESUMO

Acute respiratory distress syndrome (ARDS) is characterized by an excessive acute inflammatory response in lung parenchyma, which ultimately leads to refractory hypoxemia. One of the earliest abnormalities seen in lung injury is the elevated levels of inflammatory cytokines, among them, the soluble tumor necrosis factor (TNF-α) has a key role, which exerts cytotoxicity in epithelial and endothelial cells thus exacerbates edema. The bacterial lipopolysaccharide (LPS) was used both in vitro (RAW 264.7, THP-1, MLE-12, A549, and BEAS-2B) and in vivo (C57BL/6 mice), as it activates a plethora of overlapping inflammatory signaling pathways involved in ARDS. Nimbolide is a chemical constituent of Azadirachta indica, which contains multiple biological properties, while its role in ARDS is elusive. Herein, we have investigated the protective effects of nimbolide in abrogating the complications associated with ARDS. We showed that nimbolide markedly suppressed the nitrosative-oxidative stress, inflammatory cytokines, and chemokines expression by suppressing iNOS, myeloperoxidase, and nitrotyrosine expression. Moreover, nimbolide mitigated the migration of neutrophils and mast cells whilst normalizing the LPS-induced hypothermia. Also, nimbolide modulated the expression of epigenetic regulators with multiple HDAC inhibitory activity by suppressing the nuclear translocation of NF-κB and HDAC-3. We extended our studies using molecular docking studies, which demonstrated a strong interaction between nimbolide and TNF-α. Additionally, we showed that treatment with nimbolide increased GSH, Nrf-2, SOD-1, and HO-1 protein expression; concomitantly abrogated the LPS-triggered TNF-α, p38 MAPK, mTOR, and GSK-3ß protein expression. Collectively, these results indicate that TNF-α-regulated NF-κB and HDAC-3 crosstalk was ameliorated by nimbolide with promising anti-nitrosative, antioxidant, and anti-inflammatory properties in LPS-induced ARDS.


Assuntos
Azadirachta/química , Limoninas/uso terapêutico , Simulação de Acoplamento Molecular/métodos , Síndrome do Desconforto Respiratório/tratamento farmacológico , Síndrome do Desconforto Respiratório/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Humanos , Limoninas/farmacologia , Masculino , Camundongos , NF-kappa B/metabolismo , Síndrome do Desconforto Respiratório/patologia , Translocação Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...