Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Autophagy ; 20(6): 1359-1382, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38447939

RESUMO

Mitophagy involves the selective elimination of defective mitochondria during chemotherapeutic stress to maintain mitochondrial homeostasis and sustain cancer growth. Here, we showed that CLU (clusterin) is localized to mitochondria to induce mitophagy controlling mitochondrial damage in oral cancer cells. Moreover, overexpression and knockdown of CLU establish its mitophagy-specific role, where CLU acts as an adaptor protein that coordinately interacts with BAX and LC3 recruiting autophagic machinery around damaged mitochondria in response to cisplatin treatment. Interestingly, CLU triggers class III phosphatidylinositol 3-kinase (PtdIns3K) activity around damaged mitochondria, and inhibition of mitophagic flux causes the accumulation of excessive mitophagosomes resulting in reactive oxygen species (ROS)-dependent apoptosis during cisplatin treatment in oral cancer cells. In parallel, we determined that PPARGC1A/PGC1α (PPARG coactivator 1 alpha) activates mitochondrial biogenesis during CLU-induced mitophagy to maintain the mitochondrial pool. Intriguingly, PPARGC1A inhibition through small interfering RNA (siPPARGC1A) and pharmacological inhibitor (SR-18292) treatment counteracts CLU-dependent cytoprotection leading to mitophagy-associated cell death. Furthermore, co-treatment of SR-18292 with cisplatin synergistically suppresses tumor growth in oral cancer xenograft models. In conclusion, CLU and PPARGC1A are essential for sustained cancer cell growth by activating mitophagy and mitochondrial biogenesis, respectively, and their inhibition could provide better therapeutic benefits against oral cancer.


Assuntos
Sobrevivência Celular , Clusterina , Mitocôndrias , Mitofagia , Neoplasias Bucais , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Humanos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Clusterina/metabolismo , Clusterina/genética , Mitofagia/efeitos dos fármacos , Mitofagia/fisiologia , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Neoplasias Bucais/patologia , Neoplasias Bucais/metabolismo , Neoplasias Bucais/genética , Animais , Sobrevivência Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Biogênese de Organelas , Camundongos , Apoptose/efeitos dos fármacos , Camundongos Nus , Espécies Reativas de Oxigênio/metabolismo , Autofagia/fisiologia , Autofagia/efeitos dos fármacos
2.
Cell Death Dis ; 14(11): 732, 2023 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-37949849

RESUMO

SIRT1 (NAD-dependent protein deacetylase sirtuin-1), a class III histone deacetylase acting as a tumor suppressor gene, is downregulated in oral cancer cells. Non-apoptotic doses of cisplatin (CDDP) downregulate SIRT1 expression advocating the mechanism of drug resistance. SIRT1 downregulation orchestrates inhibition of DNM1L-mediated mitochondrial fission, subsequently leading to the formation of hyperfused mitochondrial networks. The hyperfused mitochondrial networks preserve the release of cytochrome C (CYCS) by stabilizing the mitochondrial inner membrane cristae (formation of mitochondrial nucleoid clustering mimicking mito-bulb like structures) and reducing the generation of mitochondrial superoxide to inhibit apoptosis. Overexpression of SIRT1 reverses the mitochondrial hyperfusion by initiating DNM1L-regulated mitochondrial fission. In the overexpressed cells, inhibition of mitochondrial hyperfusion and nucleoid clustering (mito-bulbs) facilitates the cytoplasmic release of CYCS along with an enhanced generation of mitochondrial superoxide for the subsequent induction of apoptosis. Further, low-dose priming with gallic acid (GA), a bio-active SIRT1 activator, nullifies CDDP-mediated apoptosis inhibition by suppressing mitochondrial hyperfusion. In this setting, SIRT1 knockdown hinders apoptosis activation in GA-primed oral cancer cells. Similarly, SIRT1 overexpression in the CDDP resistance oral cancer-derived polyploid giant cancer cells (PGCCs) re-sensitizes the cells to apoptosis. Interestingly, synergistically treated with CDDP, GA induces apoptosis in the PGCCs by inhibiting mitochondrial hyperfusion.


Assuntos
Dinâmica Mitocondrial , Neoplasias Bucais , Humanos , Superóxidos , Sirtuína 1/genética , Sirtuína 1/metabolismo , Apoptose , Cisplatino/farmacologia , Mitomicina , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/genética
4.
ACS Appl Bio Mater ; 6(10): 4314-4325, 2023 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-37782070

RESUMO

Bacterial infection and the development of antibiotic-resistant bacteria have decreased the effectiveness of traditional antibiotic treatments for wound healing. The design of a multifunctional adhesive hydrogel with antibacterial activity, self-healing properties, and on-demand removability to promote wound healing is highly desirable. In this work, a photothermal cyclodextrin with a NO-releasing moiety has been incorporated within an oxidized sodium alginate conjugated polyacrylamide (OS@PA) hydrogel to get a photothermal NO-releasing GSNOCD-OS@PA hydrogel. Such a multifunctional hydrogel has the unique feature of combined antibacterial activity as a result of a controlled photothermal effect and NO gas release under an 808 near-infrared laser. Because of oxidized sodium alginate (OSA), the hydrogel matrix easily adheres to the skin under twisted and bent states. In vitro cytotoxicity analysis against 3T3 cells showed that the hydrogels OS@PA and GSNOCD-OS@PA are noncytotoxic under laser exposure. The temperature-induced NO release by GSNOCD-OS@PA reached 31.7 mg/L when irradiated with an 808 nm laser for 10 min. The combined photothermal therapy and NO release from GSNOCD-OS@PA effectively reduced viability of both Staphylococcus aureus (Gram-positive) and Escherichia coli (Gram-negative) to 3 and 5%, respectively. Importantly, the phototherapeutic NO-releasing platform displayed effective fibroblast proliferation in a cell scratch assay.


Assuntos
Adesivos , Hidrogéis , Camundongos , Animais , Hidrogéis/farmacologia , Antibacterianos , Cicatrização , Alginatos/farmacologia
6.
Cytokine ; 171: 156366, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37716189

RESUMO

Cytokine therapy and cytokine-mediated autophagy have been used as prominent host-directed therapy (HDT) approaches to restrain M. tb growth in the host cell. In the present study, we have dissected the anti-tubercular activity of Soybean lectin (SBL) through cytokine-mediated autophagy induction in differentiated THP-1 (dTHP-1) cells. A significant increase in IL-6 expression was observed in both uninfected and mycobacteria infected dTHP-1 cells through the P2RX7 mediated pathway via PI3K/Akt/CREB-dependent signalling after SBL treatment. Inhibition of IL-6 level using IL-6 neutralizing antibody or associated signalling significantly enhanced the mycobacterial load in SBL-treated dTHP-1 cells. Further, autocrine signalling of IL-6 through its receptor-induced Mcl-1 expression activated autophagy via JAK2/STAT3 pathway, and inhibition of this pathway affected autophagy. Finally, blocking the IL-6-regulated autophagy through NSC 33994 (a JAK2 inhibitor) or S63845 (an Mcl-1 inhibitor) led to a notable increase in intracellular mycobacterial growth in SBL-treated cells. Taken together, these results indicate that SBL interacts with P2RX7 to regulate PI3K/Akt/CREB network to release IL-6 in dTHP-1 cells. The released IL-6, in turn, activates the JAK2/STAT3/Mcl-1 pathway upon interaction with IL-6Rα to modulate autophagy that ultimately controls mycobacterial growth in macrophages.


Assuntos
Interleucina-6 , Mycobacterium tuberculosis , Autofagia , Interleucina-6/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/metabolismo , Células THP-1 , Humanos
7.
Free Radic Biol Med ; 207: 72-88, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37423560

RESUMO

Cancer stem cell (CSC) populations are regulated by autophagy, which in turn modulates tumorigenicity and malignancy. In this study, we demonstrated that cisplatin treatment enriches the CSCs population by increasing autophagosome formation and speeding up autophagosome-lysosome fusion by recruiting RAB7 to autolysosomes. Further, cisplatin treatment stimulates lysosomal activity and increases autophagic flux in oral CD44+ cells. Interestingly, both ATG5- and BECN1-dependent autophagy are essential for maintaining cancer stemness, self-renewal, and resistance to cisplatin-induced cytotoxicity in oral CD44+ cells. Moreover, we discovered that autophagy-deficient (shATG5 and/or shBECN1) CD44+ cells activates nuclear factor, erythroid 2 like 2 (NRF2) signaling, which in turn reduces the elevated reactive oxygen species (ROS) level enhancing cancer stemness. Genetic inhibition of NRF2 (siNRF2) in autophagy-deficient CD44+ cells increases mitochondrial ROS (mtROS) level, reducing cisplatin-resistance CSCs, and pre-treatment with mitoTEMPO [a mitochondria-targeted superoxide dismutase (SOD) mimetic] lessened the cytotoxic effect enhancing cancer stemness. We also found that inhibiting autophagy (with CQ) and NRF2 signaling (with ML-385) combinedly increases cisplatin cytotoxicity, thereby suppressing the expansion of oral CD44+ cells; this finding has the potential to be clinically applicable in resolving CSC-associated chemoresistance and tumor relapse in oral cancer.


Assuntos
Autofagia , Cisplatino , Mitocôndrias , Neoplasias , Apoptose , Cisplatino/farmacologia , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo
8.
Drug Discov Today ; 28(9): 103692, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37379905

RESUMO

Cellular localization and deacetylation activity of sirtuin 1 (SIRT1) has a significant role in cancer regulation. The multifactorial role of SIRT1 in autophagy regulates several cancer-associated cellular phenotypes, aiding cellular survival and cell death induction. SIRT1-mediated deacetylation of autophagy-related genes (ATGs) and associated signaling mediators control carcinogenesis. The hyperactivation of bulk autophagy, disrupted lysosomal and mitochondrial biogenesis, and excessive mitophagy are key mechanism for SIRT1-mediated autophagic cell death (ACD). In terms of the SIRT1-ACD nexus, identifying SIRT1-activating small molecules and understanding the possible mechanism triggering ACD could be a potential therapeutic avenue for cancer prevention. In this review, we provide an update on the structural and functional intricacy of SIRT1 and SIRT1-mediated autophagy activation as an alternative cell death modality for cancer prevention.


Assuntos
Morte Celular Autofágica , Neoplasias , Sirtuína 1/genética , Sirtuína 1/metabolismo , Transdução de Sinais , Autofagia/genética , Neoplasias/prevenção & controle
9.
J Cell Sci ; 136(13)2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37313742

RESUMO

MTP18 (also known as MTFP1), an inner mitochondrial membrane protein, plays a vital role in maintaining mitochondrial morphology by regulating mitochondrial fission. Here, we found that MTP18 functions as a mitophagy receptor that targets dysfunctional mitochondria into autophagosomes for elimination. Interestingly, MTP18 interacts with members of the LC3 (also known as MAP1LC3) family through its LC3-interacting region (LIR) to induce mitochondrial autophagy. Mutation in the LIR motif (mLIR) inhibited that interaction, thus suppressing mitophagy. Moreover, Parkin or PINK1 deficiency abrogated mitophagy in MTP18-overexpressing human oral cancer-derived FaDu cells. Upon exposure to the mitochondrial oxidative phosphorylation uncoupler CCCP, MTP18[mLIR]-FaDu cells showed decreased TOM20 levels without affecting COX IV levels. Conversely, loss of Parkin or PINK1 resulted in inhibition of TOM20 and COX IV degradation in MTP18[mLIR]-FaDu cells exposed to CCCP, establishing Parkin-mediated proteasomal degradation of outer mitochondrial membrane as essential for effective mitophagy. We also found that MTP18 provides a survival advantage to oral cancer cells exposed to cellular stress and that inhibition of MTP18-dependent mitophagy induced cell death in oral cancer cells. These findings demonstrate that MTP18 is a novel mitophagy receptor and that MTP18-dependent mitophagy has pathophysiologic implications for oral cancer progression, indicating inhibition of MTP18-mitophagy could thus be a promising cancer therapy strategy.


Assuntos
Membranas Mitocondriais , Neoplasias Bucais , Humanos , Apoptose/genética , Carbonil Cianeto m-Clorofenil Hidrazona/metabolismo , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Dinâmica Mitocondrial , Membranas Mitocondriais/metabolismo , Mitofagia/genética , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
10.
Autophagy ; 19(8): 2196-2216, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36779631

RESUMO

Mitophagy regulates cancer stem cell (CSC) populations affecting tumorigenicity and malignancy in various cancer types. Here, we report that cisplatin treatment led to the activation of higher mitophagy through regulating CLU (clusterin) levels in oral CSCs. Moreover, both the gain-of-function and loss-of-function of CLU indicated its mitophagy-specific role in clearing damaged mitochondria. CLU also regulates mitochondrial fission by activating the Ser/Thr kinase AKT, which triggered phosphorylation of DNM1L/Drp1 at the serine 616 residue initiating mitochondrial fission. More importantly, we also demonstrated that CLU-mediated mitophagy positively regulates oral CSCs through mitophagic degradation of MSX2 (msh homeobox 2), preventing its nuclear translocation from suppressing SOX2 activity and subsequent inhibition of cancer stemness and self-renewal ability. However, CLU knockdown disturbed mitochondrial metabolism generating excessive mitochondrial superoxide, which improves the sensitivity to cisplatin in oral CSCs. Notably, our results showed that CLU-mediated cytoprotection relies on SOX2 expression. SOX2 inhibition through genetic (shSOX2) and pharmacological (KRX-0401) strategies reverses CLU-mediated cytoprotection, sensitizing oral CSCs toward cisplatin-mediated cell death.


Assuntos
Neoplasias , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-akt/metabolismo , Clusterina/genética , Clusterina/metabolismo , Cisplatino/farmacologia , Autofagia , Células-Tronco Neoplásicas/metabolismo , Dinâmica Mitocondrial/genética , Neoplasias/metabolismo
11.
J Cell Physiol ; 238(2): 287-305, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36502521

RESUMO

Recent developments in lysosome biology have transformed our view of lysosomes from static garbage disposals that can also act as suicide bags to decidedly dynamic multirole adaptive operators of cellular homeostasis. Lysosome-governed signaling pathways, proteins, and transcription factors equilibrate the rate of catabolism and anabolism (autophagy to lysosomal biogenesis and metabolite pool maintenance) by sensing cellular metabolic status. Lysosomes also interact with other organelles by establishing contact sites through which they exchange cellular contents. Lysosomal function is critically assessed by lysosomal positioning and motility for cellular adaptation. In this setting, mechanistic target of rapamycin kinase (MTOR) is the chief architect of lysosomal signaling to control cellular homeostasis. Notably, lysosomes can orchestrate explicit cell death mechanisms, such as autophagic cell death and lysosomal membrane permeabilization-associated regulated necrotic cell death, to maintain cellular homeostasis. These lines of evidence emphasize that the lysosomes serve as a central signaling hub for cellular homeostasis.


Assuntos
Apoptose , Transdução de Sinais , Humanos , Sobrevivência Celular , Homeostase/fisiologia , Transdução de Sinais/fisiologia , Lisossomos/metabolismo , Autofagia/fisiologia
13.
Semin Cancer Biol ; 80: 205-217, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-32450139

RESUMO

Autophagy is an intracellular catabolic self-cannibalism that eliminates dysfunctional cytoplasmic cargos by the fusion of cargo-containing autophagosomes with lysosomes to maintain cyto-homeostasis. Autophagy sustains a dynamic interlink between cytoprotective and cytostatic function during malignant transformation in a context-dependent manner. The antioxidant and immunomodulatory phyto-products govern autophagy and autophagy-associated signaling pathways to combat cellular incompetence during malignant transformation. Moreover, in a close cellular signaling circuit, autophagy regulates aberrant epigenetic modulation and inflammation, which limits tumor metastasis. Thus, manipulating autophagy for induction of cell death and associated regulatory phenomena will embark on a new strategy for tumor suppression with wide therapeutic implications. Despite the prodigious availability of lead pharmacophores in nature, the central autophagy regulating entities, their explicit target, as well as pre-clinical and clinical assessment remains a major question to be answered. In addition to this, the stage-specific regulation of autophagy and mode of action with natural products in regulating the key autophagic molecules, control of tumor-specific pathways in relation to modulation of autophagic network specify therapeutic target in caner. Moreover, the molecular pathway specificity and enhanced efficacy of the pre-existing chemotherapeutic agents in co-treatment with these phytochemicals hold high prevalence for target specific cancer therapeutics. Hence, the multi-specific role of phytochemicals in a cellular and tumor context dependent manner raises immense curiosity for investigating of novel therapeutic avenues. In this perspective, this review discusses about diverse implicit mechanisms deployed by the bioactive compounds in diagnosis and therapeutics approach during cancer progression with special insight into autophagic regulation.


Assuntos
Antineoplásicos , Neoplasias , Antineoplásicos/uso terapêutico , Autofagia , Transformação Celular Neoplásica/metabolismo , Humanos , Lisossomos/patologia , Neoplasias/patologia , Compostos Fitoquímicos/farmacologia , Compostos Fitoquímicos/uso terapêutico
14.
Drug Discov Today ; 27(1): 269-279, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34400351

RESUMO

Histone deacetylases (HDACs) inhibit the acetylation of crucial autophagy genes, thereby deregulating autophagy and autophagic cell death (ACD) and facilitating cancer cell survival. Vorinostat, a broad-spectrum pan-HDAC inhibitor, inhibits the deacetylation of key autophagic markers and thus interferes with ACD. Vorinostat-regulated ACD can have an autophagy-mediated, -associated or -dependent mechanism depending on the involvement of apoptosis. Molecular insights revealed that hyperactivation of the PIK3C3/VPS34-BECN1 complex increases lysosomal disparity and enhances mitophagy. These changes are followed by reduced mitochondrial biogenesis and by secondary signals that enable superactivated, nonselective or bulk autophagy, leading to ACD. Although the evidence is limited, this review focuses on molecular insights into vorinostat-regulated ACD and describes critical concepts for clinical translation.


Assuntos
Morte Celular Autofágica , Autofagia , Neoplasias , Vorinostat/farmacologia , Morte Celular Autofágica/efeitos dos fármacos , Morte Celular Autofágica/fisiologia , Autofagia/efeitos dos fármacos , Autofagia/genética , Inibidores de Histona Desacetilases/farmacologia , Humanos , Neoplasias/metabolismo , Neoplasias/prevenção & controle , Biogênese de Organelas
15.
Eur J Pharmacol ; 906: 174235, 2021 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-34097884

RESUMO

P2X7 receptor, a purinergic receptor family member, is abundantly expressed on many cells, including immune, muscle, bone, neuron, and glia. It acts as an ATP-activated cation channel that permits the influx of Ca2+, Na+ and efflux of K+ ions. The P2X7 receptor plays crucial roles in many physiological processes including cytokine and chemokine secretion, NLRP3 inflammasome activation, cellular growth and differentiation, locomotion, wound healing, transcription factors activation, cell death and T-lymphocyte survival. Past studies have demonstrated the up-regulation and direct association of this receptor in many pathophysiological conditions such as cancer, diabetics, arthritis, tuberculosis (TB) and inflammatory diseases. Hence, targeting this receptor is considered a worthwhile approach to lessen the afflictions associated with the disorders mentioned above by understanding the receptor architecture and downstream signalling processes. Here, in the present review, we have dissected the structural and functional aspects of the P2X7 receptor, emphasizing its role in various diseased conditions. This information will provide in-depth knowledge about the receptor and help to develop apt curative methodologies for the betterment of humanity in the coming years.


Assuntos
Agonistas do Receptor Purinérgico P2X/uso terapêutico , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X7/metabolismo , Animais , Artrite/tratamento farmacológico , Artrite/imunologia , Citocinas/metabolismo , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/imunologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Inflamassomos/efeitos dos fármacos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Inflamação/tratamento farmacológico , Inflamação/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Agonistas do Receptor Purinérgico P2X/farmacologia , Antagonistas do Receptor Purinérgico P2X/uso terapêutico , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Tuberculose/tratamento farmacológico , Tuberculose/imunologia
17.
ACS Appl Bio Mater ; 4(9): 7059-7069, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-35006938

RESUMO

Uniform monodispersed nitrogen-doped carbon spheres have been emerging as an exciting platform for multipurpose medical applications like photothermal therapy and photoacoustic imaging and as carriers for aromatic anticancer drugs. However, synthesis of uniform N-doped mesoporous carbon of size less than 100 nm with reasonable photothermal and photodynamic activities is a challenging task. In this connection, the present paper reports synthesis of nitrogen-doped mesoporous carbon spheres (NMCSs) from five different copolymers of pyrrole and substituted aniline (-H, o-NH2, m-NH2, p-NH2, and m-NO2) using a soft template approach. It has been found that NMCSs synthesized from poly(pyrrole-co-m-nitroaniline) show uniform mesoporous particles of size 80 nm, a photothermal conversion efficiency η of 52.7%, and an average 1O2 quantum yield of 20% under exposure of a 980 nm NIR laser. With a high η of 52%, a multifunctional nanodrug has been formulated by loading 5-Fu in NMCS. The overall drug-loaded NMC was encapsulated by thermosensitive DSPE-PEG to improve translocation of the particle in the cell and thermosensitive drug release. A reliable release of anticancer drug 5-Fu (78%) has been achieved in 50 h in lysosomal conditions under 980 nm laser exposure. This NMC-5-Fu-DSPE-PEG nanodrug produces reactive oxygen species and enhances the therapeutic effect in comparison with free drug under an NIR laser as verified in B16F0 melanoma cells.


Assuntos
Antineoplásicos , Neoplasias Cutâneas , Antineoplásicos/farmacologia , Cápsulas , Carbono , Fluoruracila , Humanos , Nitrogênio , Fosfatidiletanolaminas , Pirróis/farmacologia , Neoplasias Cutâneas/tratamento farmacológico
18.
Biochim Biophys Acta Gen Subj ; 1865(2): 129806, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33253803

RESUMO

BACKGROUND: Host-directed therapy is considered a novel anti-tuberculosis strategy in tackling the tuberculosis burden through autophagy induction by various inducers to curtail the growth of intracellular Mycobacterium tuberculosis. METHODS: In this study, we investigated the anti-tubercular role of soybean lectin, a lectin isolated from Glycine max (Soybean). Effect of SBL on intracellular mycobacterial viability through autophagy and the mechanism involved in differentiated THP-1 cells was studied using different experimental approaches. RESULTS: We initially performed a time kinetic experiment with the non-cytotoxic dose of SBL (20 µg/ml) and observed autophagy induction after 24 h of treatment. Abrogation of autophagy in the presence of 3-MA and an increase in LC3 puncta formation upon Baf-A1 addition elucidated the specific effect on autophagy and autophagic flux. SBL treatment also led to autophagy induction in mycobacteria infected macrophages that restricted the intracellular mycobacterial growth, thus emphasizing the host defensive role of SBL induced autophagy. Mechanistic studies revealed an increase in P2RX7 expression, NF-κB activation and reactive oxygen species generation upon SBL treatment. Inhibition of P2RX7 expression suppressed NF-κB dependent ROS level in SBL treated cells. Moreover, SBL induced autophagy was abrogated in the presence of either different inhibitors or P2RX7 siRNA, leading to the reduced killing of intracellular mycobacteria. CONCLUSION: Taken together, these results conclude that SBL induced autophagy exerts an anti-mycobacterial effect in P2RX7-NF-κB dependent manner through the generation of ROS. GENERAL SIGNIFICANCE: This study has provided a novel anti-mycobacterial role of SBL, which may play an important role in devising new therapeutic interventions.


Assuntos
Antibacterianos/farmacologia , Mycobacterium/efeitos dos fármacos , NF-kappa B/metabolismo , Lectinas de Plantas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas de Soja/farmacologia , Antibacterianos/isolamento & purificação , Antituberculosos/isolamento & purificação , Antituberculosos/farmacologia , Autofagia/efeitos dos fármacos , Linhagem Celular , Humanos , Macrófagos/microbiologia , Modelos Moleculares , Mycobacterium/fisiologia , Infecções por Mycobacterium/tratamento farmacológico , Infecções por Mycobacterium/metabolismo , Infecções por Mycobacterium/microbiologia , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/fisiologia , Lectinas de Plantas/isolamento & purificação , Espécies Reativas de Oxigênio/metabolismo , Proteínas de Soja/isolamento & purificação , Glycine max/química , Tuberculose/tratamento farmacológico , Tuberculose/metabolismo , Tuberculose/microbiologia
19.
Mol Biol Rep ; 47(12): 9567-9578, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33241447

RESUMO

Marine algae are an auspicious source of innovative bioactive compounds containing possible therapeutic agents against mammalian cancers. However, the mechanism by which bioactive algal compounds exhibit anticancer activity against oral squamous cell carcinoma (OSCC) is scant. The main objective of the current study was to explore the properties of the Enteromorpha compressa solvent extracts that induced autophagy and apoptosis with reference to their potent phytochemical and antioxidant properties. The presence of bioactive compounds were confirmed by UV and FT-IR spectroscopy. The free radical scavenging activity were analyzed by evaluating H2O2, DPPH, superoxide and hydroxyl activity. The anticancer activities of the extracts were investigated by employing clonogenic and scratch assay. The apoptosis potential was evaluated by DAPI and MMP by Rh123 fluorescence assay. Moreover, the CAT, SOD, GPX, APX, and GR activities were measured. The autophagy potential was evaluated by LC3 puncta formation, acridine orange in addition to LysoTracker staining. The present investigation revealed that the methanolic extract of E. compressa elicited robust free radical scavenging activity that discerns its antiproliferative potency. Moreover, the methanolic algal extract boosted intrinsic apoptosis against OSCC by downregulating protective antioxidant enzymes. Furthermore, it also revealed induction of autophagy to promote cell death in oral cancer cells. The presence of novel bioactive compounds in E. compressa has uncovered possible therapeutic value against OSCC by modulating antioxidant defense system, apoptosis and autophagy that could be used to explore very competent algal candidates for the development of potential alternative anticancer drugs.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Ulva/química , Antineoplásicos Fitogênicos/isolamento & purificação , Antioxidantes/isolamento & purificação , Apoptose/genética , Ascorbato Peroxidases/genética , Ascorbato Peroxidases/metabolismo , Autofagia/genética , Compostos de Bifenilo/antagonistas & inibidores , Caspase 3/genética , Caspase 3/metabolismo , Caspase 7/genética , Caspase 7/metabolismo , Catalase/genética , Catalase/metabolismo , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Glutationa Redutase/genética , Glutationa Redutase/metabolismo , Humanos , Peróxido de Hidrogênio/antagonistas & inibidores , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Picratos/antagonistas & inibidores , Extratos Vegetais/química , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
20.
Int J Biol Macromol ; 164: 4263-4278, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32916197

RESUMO

Fucoidan is a complex polysaccharide (molecular weight 10,000-100,000 Da) derived from brown algae which comprises of L-fucose and sulfate groups have potential as therapeutic diligences against several human diseases. The fucoidan has expanded a widespread range of pharmacological properties as an anti-inflammatory, anticoagulant, antiangiogenic, immunomodulatory, anti-adhesive, anticancer, antidiabetic, antiviral and anti-neurodegenerative agents owing to their diverse chemical conformation and potent antioxidant activity. The antioxidant and immunomodulatory activities of the fucoidan contribute towards their disease preventive potency through dynamic modulation of key intracellular signalling pathways, regulation of ROS accumulation, and maintenance of principal cell survival and death pathways. Additionally, it also reduces cancer-associated cachexia. Despite the wide range of therapeutic potency, the fucoidan is heavily regarded as an unexplored plethora of druggable entities in the current situation. The isolation, screening, biological application, pre-clinical, and clinical assessment along with large scale cost-effective production remain a foremost task to be assessed. Moreover, the chemical synthesis of the present bioactive drug with confirmational rearrangement for enhanced availability and bioactivity also need tenacious investigation. Hence, in the present review, we give attention to the source of isolation of fucoidan, their principle strategic deployment in disease prevention, and the mechanistic investigation of how it works to combat different diseases that can be used for future therapeutic intervention.


Assuntos
Organismos Aquáticos/química , Polissacarídeos/química , Sulfatos/química , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Fracionamento Químico/métodos , Suscetibilidade a Doenças , Descoberta de Drogas , Saúde , Humanos , Imunomodulação/efeitos dos fármacos , Desintoxicação Metabólica Fase I , Estrutura Molecular , Polissacarídeos/isolamento & purificação , Alga Marinha/química , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...