Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 22(1): 189, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38519981

ABSTRACT

The proinflammatory cytokines and arachidonic acid (AA)-derived eicosanoids play a key role in cartilage degeneration in osteoarthritis (OA). The lysophosphatidylcholine acyltransferase 3 (LPCAT3) preferentially incorporates AA into the membranes. Our recent studies showed that MALT1 [mucosa-associated lymphoid tissue lymphoma translocation protein 1]) plays a crucial role in propagating inflammatory signaling triggered by IL-1ß and other inflammatory mediators in endothelial cells. The present study shows that LPCAT3 expression was up-regulated in both human and mice articular cartilage of OA, and correlated with severity of OA. The IL-1ß-induces cell death via upregulation of LPCAT3, MMP3, ADAMTS5, and eicosanoids via MALT1. Gene silencing or pharmacological inhibition of LPCAT3 or MALT1 in chondrocytes and human cartilage explants notably suppressed the IL-1ß-induced cartilage catabolism through inhibition of expression of MMP3, ADAMTS5, and also secretion of cytokines and eicosanoids. Mechanistically, overexpression of MALT1 in chondrocytes significantly upregulated the expression of LPCAT3 along with MMP3 and ADAMTS5 via c-Myc. Inhibition of c-Myc suppressed the IL-1ß-MALT1-dependent upregulation of LPCAT3, MMP3 and ADAMTS5. Consistent with the in vitro data, pharmacological inhibition of MALT1 or gene silencing of LPCAT3 using siRNA-lipid nanoparticles suppressed the synovial articular cartilage erosion, pro-inflammatory cytokines, and eicosanoids such as PGE2, LTB4, and attenuated osteoarthritis induced by the destabilization of the medial meniscus in mice. Overall, our data reveal a previously unrecognized role of the MALT1-LPCAT3 axis in osteoarthritis. Targeting the MALT1-LPCAT3 pathway with MALT1 inhibitors or siRNA-liposomes of LPCAT3 may become an effective strategy to treat OA by suppressing eicosanoids, matrix-degrading enzymes, and proinflammatory cytokines.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Humans , Mice , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/pharmacology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Cytokines/metabolism , Eicosanoids/metabolism , Eicosanoids/pharmacology , Eicosanoids/therapeutic use , Endothelial Cells/metabolism , Interleukin-1beta/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 3/pharmacology , Matrix Metalloproteinase 3/therapeutic use , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Osteoarthritis/metabolism , RNA, Small Interfering/metabolism
2.
Bioorg Chem ; 146: 107294, 2024 May.
Article in English | MEDLINE | ID: mdl-38507997

ABSTRACT

Oxindoles are potent anti-cancer agents and are also used against microbial and fungal infections and for treating neurodegenerative diseases. These oxindoles are earlier established as estrogen receptor (ER)-targeted agents for killing ER (+) cancer cells. Our previously developed bis-arylidene oxindole, Oxifen (OXF) exhibits effective targeting towards ER (+) cancer cells which has a structural resemblance with tamoxifen. Herein, we have designed and synthesized few structural analogues of OXF such as BPYOX, ACPOX and ACPOXF to examine its cytotoxicity in different cancer as well as non-cancer cell lines and its potential to form self- aggregates in aqueous solution. Among these series of molecules, ACPOXF showed maximum toxicity in colorectal cancer cell line which are ER (-) but it also kills non-cancer cell line HEK-293, thereby reducing its cancer cell selectivity. Incidentally, ACPOXF exhibits self-aggregation, without the help of a co-lipid with nanometric size in aqueous solution. ACPOXF self-aggregate was co-formulated with glucocorticoid receptor (GR) synthetic ligand, dexamethasone (Dex) (called, ACPOXF-Dex aggregate) which could selectively kill ER (-) colorectal cancer cells and also could increase survivability of colon-tumour bearing mice. ACPOXF-Dex induced ROS up-regulation followed by apoptosis through expression of caspase-3. Further, we observed upregulation of antiproliferative factor, p53 and epithelial-to-mesenchymal (EMT) reversal marker E-cadherin in tumour mass. In conclusion, a typical structural modification in ER-targeting Oxifen moiety resulted in its self-aggregation that enabled it to carry a GR-ligand, thus broadening its selective antitumor property especially as colon cancer therapeutics.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Mice , Humans , Animals , Ligands , HEK293 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Apoptosis , Receptors, Estrogen/metabolism , Oxindoles/chemistry , Colorectal Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation
3.
J Colloid Interface Sci ; 663: 82-93, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38394820

ABSTRACT

HYPOTHESIS: Lipid nanoparticles containing a cationic lipid are increasingly used in drug and gene delivery as they can display improved cellular uptake, enhanced loading for anionic cargo such as siRNA and mRNA or exhibit additional functionality such as cytotoxicity against cancer cells. This research study tests the hypothesis that the molecular structure of the cationic lipid influences the structure of the lipid nanoparticle, the cellular uptake, and the resultant cytotoxicity. EXPERIMENTS: Three potentially cytotoxic cationic lipids, with systematic variations to the hydrophobic moiety, were designed and synthesised. All the three cationic lipids synthesised contain pharmacophores such as the bicyclic coumarin group (CCA12), the tricyclic etodolac moiety (ETD12), or the large pentacyclic triterpenoid "ursolic" group (U12) conjugated to a quaternary ammonium cationic lipid containing twin C12 chains. The cationic lipids were doped into monoolein cubosomes at a range of concentrations from 0.1 mol% to 5 mol% and the effect of the lipid molecular architecture on the cubosome phase behaviour was assessed using a combination of Small Angle X-Ray Scattering (SAXS), Dynamic Light Scattering (DLS), zeta-potential and cryo-Transmission Electron Microscopy (Cryo-TEM). The resulting cytotoxicity of these particles against a range of cancerous and non-cancerous cell-lines was assessed, along with their cellular uptake. FINDINGS: The molecular architecture of the cationic lipid was linked to the internal nanostructure of the resulting cationic cubosomes with a transition to more curved cubic and hexagonal phases generally observed. Cubosomes formed from the cationic lipid CCA12 were found to have improved cellular uptake and significantly higher cytotoxicity than the cationic lipids ETD12 and U12 against the gastric cancer cell-line (AGS) at lipid concentrations ≥ 75 µg/mL. CCA12 cationic cubosomes also displayed reasonable cytotoxicity against the prostate cancer PC-3 cell-line at lipid concentrations ≥ 100 µg/mL. In contrast, 2.5 mol% ETD12 and 2.5 mol% U12 cubosomes were generally non-toxic against both cancerous and non-cancerous cell lines over the entire concentration range tested. The molecular architecture of the cationic lipid was found to influence the cubosome phase behaviour, the cellular uptake and the toxicity although further studies are necessary to determine the exact relationship between structure and cellular uptake across a range of cell lines.


Subject(s)
Nanoparticles , Scattering, Small Angle , X-Ray Diffraction , Nanoparticles/chemistry , Microscopy, Electron, Transmission , Dynamic Light Scattering , Molecular Structure
4.
Biomed Mater ; 19(2)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38290150

ABSTRACT

Glucocorticoid and Mineralocorticoid receptors are principally ligand-dependent intracellular transcription factors that are known to influence the development and growth of many human cancers. Our study investigates the potential of these receptors to act as a target for oral cancer treatment since findings in this regard are sparse till date. Leveraging the aberrant behavior of steroid hormone receptors (SHRs) in cancer, we have targeted oral cancer cells in 2D-culture using liposomes containing both synthetic as well as crude, natural SHR ligands isolated from an aqueous Indian medicinal plant. Lipoplexes thus formulated demonstrated targeted transfectability as indicated by expression of green fluorescent protein. Transfection of oral squamous cell carcinoma cells with exogenous, anticancer gene p53 lipoplexed with crude saponin-based liposome induced apoptosis of cancer cells via regulation of BAX and B-cell leukemia/lymphoma-2 (BCL2) protein levels at levels comparable with pre-established delivery systems based on synthetic SHR ligands. Our findings strongly indicate a possibility of developing plant saponin-based inexpensive delivery systems which would target cancer cells selectively with reduced risks of off target delivery and its side effects.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Saponins , Humans , Mouth Neoplasms/therapy , Transfection , Liposomes , Hormones , Steroids
5.
J Photochem Photobiol B ; 251: 112846, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38237432

ABSTRACT

Photodynamic therapy (PDT) is a clinically approved, non-invasive alternate cancer therapy. A synthetic glucocorticoid (GC), dexamethasone (Dex) has previously been demonstrated to sensitize cancer cells to chemotherapy. However, to the best of our knowledge, the sensitization effect of GCs on PDT has not yet been investigated. We hypothesized that glucocorticoid receptor (GR) targeting can selectively make cancer cells more sensitive to PDT treatment, as PDT induces hypoxia wherein GR-activity gets enhanced. In addition, Dex was reported to act against the PDT-induced cell survival pathways like HIF-1α, NRF2, NF-κB, STAT3 etc. Thus, both the treatments can complement each other and may result in increasing the effectiveness of combination therapy. Hence, in this study, we developed liposomal formulations of our previously reported PDT agent P-Nap, either alone (D1P-Nap) or in combination with Dex (D1XP-Nap) to elucidate the sensitization effect. Interestingly, our RT-PCR results in hypoxic conditions showed down-regulation of HIF-1α and over expression of GR-activated genes for glucose-6-phosphatase (G6Pase) and PEPCK enzymes, indicating prominent GR-transactivation. We also observed higher phototoxicity in CT26.WT cells treated with D1XP-Nap PDT under hypoxic conditions as compared to normoxic conditions. These effects were reversed when cells were pre-treated with RU486, a competitive inhibitor of GCs. Moreover, our in vivo findings of subcutaneous tumor model of Balb/C mice for colon cancer revealed a significant decrease in tumor volume as well as considerable enhancement in the survivability of PDT treated tumor-bearing mice when Dex was present in the formulation. A high Bax/Bcl-xL ratio, high p53 expression, enhanced E-cadherin expression and down-regulation of pro-tumorigenic transcription factors NF-κB and c-Myc were found in tumor lysates from mice treated with D1XP-Nap under PDT, indicating GR-mediated sensitization of the tumor to PDT-induced cell death and enhancement of life-span for tumor bearing mice.


Subject(s)
Colonic Neoplasms , Photochemotherapy , Mice , Animals , Receptors, Glucocorticoid/metabolism , NF-kappa B , Cell Death , Colonic Neoplasms/drug therapy , Cell Line, Tumor
7.
Biomed Mater ; 18(5)2023 08 02.
Article in English | MEDLINE | ID: mdl-37467764

ABSTRACT

Folate receptor (FR) (α) has long been the subject of active interest as regards its potential to serve as a target for cancer therapy. FR has been found to be overexpressed in several cancers, including clinical samples of different stages from OSCC (oral squamous cell carcinoma) patients. However, no clear correlation or conclusive finding has been obtained so far which might indicate the efficacy of FR as a credible target for the treatment of OSCC. All cell lines to be used were assessed for FR-expression. Subsequently, we developed glucose-derived carbon nanospheres (CSPs) and primed them with a Folate-based cationic lipid FA8 and the chemotherapeutic drug doxorubicin (DOX). CSP based delivery systems along with pristine drug DOX were characterized and treated subsequently toin vitrocultures of OSCC cells and assessed for cancer cell targetability as well as cell death. Subsequently, treatment was administered to immunocompetent C57 mice carrying MOC2 based syngeneic OSCC tumours and assessed for tumour regression and toxicity. Ligand primed targeted CSPs exhibited commendable drug uptake as well as efficient induction of cell death. Further, receptor blocking studies revealed FR-mediated uptake, preferentially in cancer cells. Drug once delivered by ligand-primed CSPs was retained longer inside cells than pristine drug alone, indicating possibilities of better therapeutic outcome. In animal studies, CSP-FA8-DOX (Ligand primed targeted CSP) demonstrated significant regression in tumour size compared to pristine DOX as well as CSP-DOX (non-targeted CSP) treated animals. FR-mediated system CFD demonstrated targeted drug uptake and apoptotic death selectively in cancer cells. Significant tumour regression was also observedin vivo. Overall, it may be presumed that the FR is a therapeutic target with substantial potential in OSCC treatment.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Nanospheres , Mice , Animals , Ligands , Carcinoma, Squamous Cell/drug therapy , Mouth Neoplasms/drug therapy , Drug Delivery Systems , Doxorubicin/pharmacology , Cell Line, Tumor , Folic Acid/metabolism
8.
J Colloid Interface Sci ; 649: 966-976, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37392686

ABSTRACT

Hypothesis The study aimed to use molecular hybridization of a cationic lipid with a known pharmacophore to produce a bifunctional lipid having a cationic charge to enhance fusion with the cancer cell surface and biological activity via the pharmacophoric head group. Experiments The novel cationic lipid DMP12 [N-(2-(3-(3,4-dimethoxyphenyl) propanamido) ethyl)-N-dodecyl-N-methyldodecan-1-aminium iodide] was synthesised by conjugating 3-(3,4-dimethoxyphenyl) propanoic acid (or 3,4-dimethoxyhydrocinnamic acid) to twin 12 carbon chains bearing a quaternary ammonium group [N-(2-aminoethyl)-N-dodecyl-N-methyldodecan-1-aminium iodide]. The physicochemical and biological properties of DMP12 were investigated. Cubosome particles consisting of monoolein (MO) doped with DMP12 and paclitaxel were characterized using Small-angle X-ray Scattering (SAXS), Dynamic Light Scattering (DLS), and Cryo-Transmission Electron Microscopy (Cryo-TEM). Combination therapy using these cubosomes was assessed in vitro against the gastric (AGS) and prostate (DU-145 and PC-3) cancer cell lines using cytotoxicity assay. Findings Monoolein (MO) cubosomes doped with DMP12 were observed to be toxic against the AGS and DU-145 cell-lines at higher cubosome concentrations (≥100 µg/ml) but had limited activity against the PC-3 cell-line. However, combination therapy consisting of 5 mol% DMP12 and 0.5 mol% paclitaxel (PTX) significantly increased the cytotoxicity against the PC-3 cell-line which was resistant to either DMP12 or PTX individually. The results demonstrate that DMP12 has a prospective role as a bioactive excipient in cancer therapy.


Subject(s)
Antineoplastic Agents , Nanoparticles , Prostatic Neoplasms , Humans , Male , Paclitaxel/chemistry , Scattering, Small Angle , Iodides , X-Ray Diffraction , Cell Line
9.
Mol Pharm ; 20(6): 2822-2835, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37134112

ABSTRACT

Progesterone (PR) is an endogenous steroid hormone that activates the progesterone receptor (PgR) and is known to play a critical role in cancer progression. Herein, we report the development of cationic lipid-conjugated PR derivatives by covalently conjugating progesterone with cationic lipids of varying hydrocarbon chain lengths (n = 6-18) through a succinate linker. Cytotoxicity studies performed on eight different cancer cell lines reveal that PR10, one of the lead derivatives, exerts notable toxicity (IC50 = 4-12 µM) in cancer cells irrespective of their PgR expression status and remains largely nontoxic to noncancerous cells. Mechanistic studies show that PR10 induces G2/M-phase cell cycle arrest in cancer cells, leading to apoptosis and cell death by inhibiting the PI3K/AKT cell survival pathway and p53 upregulation. Further, in vivo study shows that PR10 treatment significantly reduces melanoma tumor growth and prolongs the overall survival of melanoma tumor-bearing C57BL/6J mice. Interestingly, PR10 readily forms stable self-aggregates of ∼190 nm size in an aqueous environment and exhibits selective uptake into cancerous cell lines. In vitro uptake mechanism studies in various cell lines (cancerous cell lines B16F10, MCF7, PC3, and noncancerous cell line HEK293) using endocytosis inhibition proves that PR10 nanoaggregates enter selectively into the cancer cells predominantly using macropinocytosis and/or caveolae-mediated endocytosis. Overall, this study highlights the development of a self-aggregating cationic derivative of progesterone with anticancer activity, and its cancer cell-selective accumulation in nanoaggregate form holds great potential in the field of targeted drug delivery.


Subject(s)
Melanoma , Progesterone , Mice , Animals , Humans , Progesterone/pharmacology , Cell Line, Tumor , Phosphatidylinositol 3-Kinases/metabolism , HEK293 Cells , Mice, Inbred C57BL , Apoptosis , Melanoma/drug therapy , Lipids/pharmacology , Cell Proliferation
10.
Chembiochem ; 24(12): e202200650, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36905093

ABSTRACT

Drug resistance has a major impact on the treatment of several cancers. This is mainly due to the overexpression of cellular drug efflux proteins. Hence, drug-delivery systems that can avoid this resistance are needed. We report PR10, a progesterone-cationic lipid conjugate, as a self-assembling nanoaggregate that delivers a drug cargo of etoposide, a topoisomerase inhibitor, selectively to cancer cells. In this study, we observed that etoposide nanoaggregates (P : E) caused selective and enhanced toxicity in etoposide-resistant CT26 cancer cells (IC50 9 µM) compared to when etoposide (IC50 >20 µM) was used alone. Concurrently, no toxicity was observed in etoposide-sensitive HEK293 cells for P : E treatment (IC50 >20 µM). The P : E-treated cancer cells seem to have no effect on ABCB1 expression, but etoposide-treated cells exhibited a twofold increase in ABCB1 expression, a potent efflux protein for several xenobiotic compounds. This observation supports the notion that the enhanced toxicity of P : E nanoaggregates is due to their ability to keep the expression of ABCB1 low, thus allowing longer intracellular residence of etoposide. In a BALB/c orthotopic colorectal cancer model, the nanoaggregates led to enhanced survival (45 days) compared to etoposide-treated mice (39 days). These findings suggest that PR10 could be used as a potential cancer-selective etoposide delivery vehicle to treat several etoposide-resistant cancers with fewer side effects due to the nonspecific toxicity of the drug.


Subject(s)
Colorectal Neoplasms , Progesterone , Mice , Humans , Animals , Etoposide/pharmacology , Etoposide/therapeutic use , Etoposide/metabolism , HEK293 Cells , Colorectal Neoplasms/drug therapy , Lipids
11.
Bioorg Chem ; 133: 106395, 2023 04.
Article in English | MEDLINE | ID: mdl-36753964

ABSTRACT

Breast cancer is a heterogeneous malignancy with wide-ranging variations in therapeutic responses, overall survival etc. Major challenges for available chemotherapeutic agents in achieving clinical success are in maintaining systemic bio-distribution and avoiding non-specific adverse effects. Bis-arylidene oxindoles are estrogen receptor (ER)-selective bioactive molecules with moderate potency. In here, we have designed, synthesized and evaluated a series of twin aliphatic chain cationic lipid-conjugated bis-arylidene oxindole molecules with variations in nature of linker, lengths of carbon spacer and hydrophobic twin chains. We observed that among the various structural analogues, C8 twin-chain containing molecules, PGC8, S2C8 and S3C8 showed effective cancer cell-selective cytotoxicity in different cancer cell lines with an IC50 ranging from 4 to 7 µM. These molecules selectively induced apoptosis, ROS production and cell cycle inhibition at G1/S phase in ER + breast cancer cells but not in non-cancer cells. Additionally, these molecules formed homogenous self-assemblies exhibiting effective hydrodynamic diameter with positive surface charge. The self-assemblies also showed prominent cancer cell-selective uptake and DNA-binding abilities. Hence, we have shown successful incorporation of dexamethasone to the self-assemblies, and its enhanced cytotoxicity even in ER-negative breast cancer cells. All these results indicate that PGC8, S2C8 and S3C8 molecules, albeit their potent and selective ER-positive anti-breast cancer activity, can be repurposed as targeted delivery systems and hold promise as unique, broader spectrum breast cancer cell-selective therapeutic payloads.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Humans , Female , Oxindoles/pharmacology , Oxindoles/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line , Cell Cycle Checkpoints , Lipids/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Apoptosis , Cell Line, Tumor
12.
J Photochem Photobiol B ; 238: 112625, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36529058

ABSTRACT

Photodynamic therapy (PDT) is a promising non-invasive treatment modality for cancer and can be potentiated by combination with chemotherapy. Here, we combined PDT of novel porphyrin-based photosensitizers with low dose doxorubicin (Dox) to get maximum outcome. Dox potentiated and showed synergism with PDT under in vitro conditions on CT26.WT cells. The current colon cancer treatment strategies assure partial or even complete tumour regression but loco-regional relapse or distant metastasis is the major cause of death despite combination therapy. The spared cells after the treatment contribute to relapse and it is important to study their behaviour in host environment. Hence, we developed relapse models for PDT, Dox and combination treatments by transplanting respectively treated equal number of live cells to mice (n = 5) for tumour formation. Most of the treated cells lost tumour forming ability, but some treatment resistant cells developed tumours in few mice. These tumours served as relapse models and Western blot analysis of tumour samples provided clinically relevant information to delineate resistance strategies of individual as well as combination therapies at molecular level. Our results showed that low dose Dox helped in increasing the tumour inhibiting effect of PDT in combination therapy, but still there are indeed possibilities of relapse at later stages due to chemoresistance and immune suppression that may occur post-treatment. We observed that the combination therapy may also lead to the development of multidrug resistant (MDR) phenotype during relapse. Thus, this study provided clinically relevant information to further strengthen and improve PDT-drug combination therapy in order to avoid relapse and to treat cancer more effectively.


Subject(s)
Colonic Neoplasms , Photochemotherapy , Porphyrins , Mice , Animals , Porphyrins/pharmacology , Porphyrins/therapeutic use , Photochemotherapy/methods , Neoplasm Recurrence, Local/drug therapy , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Colonic Neoplasms/drug therapy , Cell Line, Tumor
13.
Chem Asian J ; 18(2): e202201136, 2023 Jan 17.
Article in English | MEDLINE | ID: mdl-36482874

ABSTRACT

Paclitaxel (PTX) is a widely used chemotherapeutic agent in the clinic. However, its clinical benefit is limited due to its low water solubility, off-target toxicity, and for being a multidrug-resistant (MDR) substrate. To overcome these limitations in this study, a tumor-targeting peptide (CRGDK peptide, a ligand for NRP-1 receptor) conjugate of α-tocopheryl succinate (α-TOS) was synthesized and modified on PTX-loaded lipid aggregate (TL-PTX) to leverage the benefits of α-TOS, which include a) anti-cancer activity, b) increased PTX loading, and c) inhibition of MDR activity. Use of peptide conjugate of α-TOS (α-TOS-CRGDK) in lipid aggregate increased PTX entrapment efficiency by 20%, helped in NRP-1 specific cellular uptake and significantly enhanced apoptotic and cell killing activity (p <0.01) of PTX compared to control formulation (CL-PTX) by inhibiting MDR-activity in melanoma resulting in ∼70% increment in overall survival of melanoma tumor-bearing mice. In conclusion, CRGDK- α-TOS conjugate in association with PTX-loaded liposome provided a unique NRP-1 targeted, drug-resistant reversing anticancer regimen for treating aggressive melanoma.


Subject(s)
Melanoma , Paclitaxel , Mice , Animals , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , alpha-Tocopherol/pharmacology , Drug Delivery Systems/methods , Peptides/pharmacology , Lipids , Cell Line, Tumor , Drug Resistance, Neoplasm
14.
Cell Biol Int ; 47(4): 742-753, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36573403

ABSTRACT

Breast cancer (BC) remains one of the deadliest and frequently diagnosed metastatic cancers worldwide. Cancer stem cells (CSCs) are the cell population within the tumor niche, having an epithelial to mesenchymal (EMT) transition phenotype, high self-renewal, vigorous metastatic capacity, drug resistance, and tumor relapse. Identification of targets for induction of apoptosis is essential to provide novel therapeutic approaches in BC. Our earlier studies showed that Vitamin C induces apoptotic cell death by losing redox balance in TNBC CSCs. In this study, we have attempted to identify previously unrecognized CSC survival factors that can be used as druggable targets for bCSCs apoptosis regulators isolated from the TNBC line, MDA MB 468. After a thorough literature review, Oct-4 was identified as the most promising marker for its unique abundance in cancer and absence in normal cells and the contribution of Oct-4 to the sustenance of cancer cells. We then validated a very high expression of Oct-4 in the MDA MB 468 bCSCs population using flow-cytometry. The loss of Oct-4 was carried out using small interfering RNA (siRNA)-mediated knockdown in the bCSCs, followed by assessing for cellular apoptosis. Our results indicated that Oct-4 knockdown induced cell death, changes in cellular morphology, inhibited mammosphere formation, and positive for Annexin-V expression, thereby indicating the role of Oct-4 in bCSC survival. Moreover, our findings also suggest the direct interaction between Oct-4 and Vitamin C using in silico docking. This data, hence, contributes towards novel information about Oct-4 highlighting this molecule as a novel survival factor in bCSCs.


Subject(s)
Breast Neoplasms , Triple Negative Breast Neoplasms , Humans , Female , Triple Negative Breast Neoplasms/metabolism , Octamer Transcription Factor-3/metabolism , Epithelial-Mesenchymal Transition , Vitamins , Neoplastic Stem Cells/metabolism , Ascorbic Acid , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Hyaluronan Receptors/metabolism
15.
ACS Macro Lett ; 11(3): 289-295, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35575367

ABSTRACT

Targeted delivery of therapeutics such as small molecule drugs or nucleic acids exclusively to the nucleus of diseased mammalian cells poses a significant challenge. The development of targeting ligands that can specifically enter certain cancer cells via a specific receptor-mediated endocytosis and then traffic exclusively to the nucleus to deliver the cargo inside it can achieve this goal. We have developed an end-functionalized shikimoylated-polypeptide with pendant shikimoyl moieties that can enter mammalian cells via the mannose receptors and are then exclusively trafficked into the nucleus. The presence of the shikimoyl group in the polypeptide, which traffics it exclusively to the nucleus, contrasts with the mannosylated or galactosylated glycopolypeptides that are distributed all over the cytoplasm or the mannose-6-phosphate containing polypeptide that is exclusively trafficked to the lysosome. Using challenge experiments, we demonstrate that these polypeptides can enter both dendritic and cancer cells through mannose-receptors and subsequently enter the cell nucleus via the interaction with a nuclear pore complex (NPC) protein importin-α/ß1. To the best of our knowledge, this represents the first example of a synthetic polyvalent glycopolypeptide mimic that performs the dual function of entering mammalian cells through specific receptors and subsequently traffics into the nucleus. The conjugation of these end-functionalized shikimoylated-polypeptides to other biological entities, such as recombinant anticancer drugs, DNA, RNA, and CRISPR-Cas9, may be a suitable alternative for delivery of these biological entities into cells affected by cancer and other genetic diseases.


Subject(s)
Cell Nucleus , Nuclear Pore Complex Proteins , Animals , Cell Nucleus/genetics , Cytoplasm/metabolism , Endocytosis , Mammals/metabolism , Nuclear Pore Complex Proteins/metabolism , Peptides/metabolism
16.
ACS Appl Mater Interfaces ; 14(13): 14859-14870, 2022 Apr 06.
Article in English | MEDLINE | ID: mdl-35347979

ABSTRACT

Long-term application of topical therapeutics for psoriasis has a plethora of side effects. Additionally, skin-permeating agents used in their formulations for deeper dermal delivery damage the skin. To address these limitations, we developed novel lithocholic acid analogues that could form lipid nanoparticles (nano-LCs) spontaneously in the aqueous milieu, permeate through the skin, penetrate the deeper dermal layers, and exert anti-inflammatory effects against psoriasis-like chronic skin inflammations. Prior findings demonstrated that lithocholic acid acts as a vitamin D receptor agonist without affecting the Ca+2 metabolism and also as an antagonist for ephrin type-A receptor 2 (EphA2). Taking cues from the previous findings, lithocholic acid derivatives with twin alkyl chains (LC6, LC8, LC10, and LC-12) were synthesized, nanoparticles (nano-LCs) were prepared, and they were evaluated for their skin permeability and anti-inflammatory properties. Among these nano-LCs, nano-LC10 demonstrated superior anti-inflammatory properties and inhibition of keratinocyte proliferation in various cell-based evaluations. Furthermore, the therapeutic efficiency of nano-LC10 was evaluated in an imiquimod-induced psoriasis-like mouse model and demonstrated comparable efficiency with the standard topical formulation, Sorvate, in reducing skin inflammations. Nano-LC10 also reduced systemic inflammation, organ toxicity, and also proinflammatory serum cytokine levels. Overall, nano-lithocholic lipidoid (nano-LC10) can be a potential novel class of therapeutics for topical application in treating psoriasis.


Subject(s)
Nanoparticles , Psoriasis , Animals , Disease Models, Animal , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Liposomes , Mice , Psoriasis/drug therapy , Psoriasis/metabolism , Skin
17.
J Med Chem ; 65(3): 1822-1834, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35019659

ABSTRACT

Herein, we report three new metal-free, photochemically active single, dual, and combinatorial CORMs (photoCORMs) based on a carbazole-fused 1,3-dioxol-2-one moiety which released one equivalent of CO, two equivalent of CO, and a combination of one equivalent of each CO and anticancer drug upon one- and two-photon excitation, respectively. The photoCORMs exhibited good cellular uptake and real-time monitoring ability of CO uncaging by a color change approach in cancerous B16F10 cells. Interestingly, the cytotoxicity assay on B16F10 cells indicated that the dual photoCORM has increased anticancer activity over the single and combinatorial photoCORMs upon irradiation. Our results also showed that CO could accelerate the effectiveness of the well-known anticancer drug (chlorambucil). Finally, the in vivo evaluation of the dual photoCORM on an established murine melanoma tumor (C57BL/6J mouse model) manifested a significant regression of tumor volume and led to significant improvement (>50%) in the overall survivability.


Subject(s)
Antineoplastic Agents/therapeutic use , Carbazoles/therapeutic use , Carbon Monoxide/metabolism , Dioxoles/therapeutic use , Melanoma/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Apoptosis/drug effects , Carbazoles/chemical synthesis , Carbazoles/radiation effects , Cell Line, Tumor , Dioxoles/chemical synthesis , Dioxoles/radiation effects , Female , Infrared Rays , Mice, Inbred C57BL , Photons
18.
Article in English | MEDLINE | ID: mdl-34541822

ABSTRACT

The steroid hormone receptors (SHRs) among nuclear hormone receptors (NHRs) are steroid ligand-dependent transcription factors that play important roles in the regulation of transcription of genes promoted via hormone responsive elements in our genome. Aberrant expression patterns and context-specific regulation of these receptors in cancer, have been routinely reported by multiple research groups. These gave an window of opportunity to target those receptors in the context of developing novel, targeted anticancer therapeutics. Besides the development of a plethora of SHR-targeting synthetic ligands and the availability of their natural, hormonal ligands, development of many SHR-targeted, anticancer nano-delivery systems and theranostics, especially based on small molecules, have been reported. It is intriguing to realize that these cytoplasmic receptors have become a hot target for cancer selective delivery. This is in spite of the fact that these receptors do not fall in the category of conventional, targetable cell surface bound or transmembrane receptors that enjoy over-expression status. Glucocorticoid receptor (GR) is one such exciting SHR that in spite of it being expressed ubiquitously in all cells, we discovered it to behave differently in cancer cells, thus making it a truly druggable target for treating cancer. This review selectively accumulates the knowledge generated in the field of SHR-targeting as a major focus for cancer treatment with various anticancer small molecules and nanotherapeutics on progesterone receptor, mineralocorticoid receptor, and androgen receptor while selectively emphasizing on GR and estrogen receptor. This review also briefly highlights lipid-modification strategy to convert ligands into SHR-targeted cancer nanotherapeutics. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures Therapeutic Approaches and Drug Discovery > Emerging Technologies.


Subject(s)
Neoplasms , Receptors, Glucocorticoid , Hormones , Humans , Ligands , Lipids , Neoplasms/drug therapy , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Steroids
19.
Chem Commun (Camb) ; 57(92): 12329-12332, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34740232

ABSTRACT

Herein, we document a self-assembling octyl-TPP tagged esculetin (Mito-Esc) as functionally active and as a novel small molecule siRNA delivery vector. While Mito-Esc itself induces selective breast cancer cell death, the amphiphilic nature of Mito-Esc delivers therapeutic siRNAs intracellularly without the need for any excipient to exacerbate the anti-proliferative effects.


Subject(s)
Mitochondria , RNA, Small Interfering , Cell Line, Tumor , Humans , Mitochondria/metabolism , RNA, Double-Stranded , Umbelliferones
20.
Molecules ; 26(15)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34361779

ABSTRACT

Delivering nucleic acids into the endothelium has great potential in treating vascular diseases. However, endothelial cells, which line the vasculature, are considered as sensitive in nature and hard to transfect. Low transfection efficacies in endothelial cells limit their potential therapeutic applications. Towards improving the transfection efficiency, we made an effort to understand the internalization of lipoplexes into the cells, which is the first and most critical step in nucleic acid transfections. In this study, we demonstrated that the transient modulation of caveolae/lipid rafts mediated endocytosis with the cholesterol-sequestrating agents, nystatin, filipin III, and siRNA against Cav-1, which significantly increased the transfection properties of cationic lipid-(2-hydroxy-N-methyl-N,N-bis(2-tetradecanamidoethyl)ethanaminium chloride), namely, amide liposomes in combination with 1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) (AD Liposomes) in liver sinusoidal endothelial cells (SK-Hep1). In particular, nystatin was found to be highly effective with 2-3-fold enhanced transfection efficacy when compared with amide liposomes in combination with Cholesterol (AC), by switching lipoplex internalization predominantly through clathrin-mediated endocytosis and macropinocytosis.


Subject(s)
Caveolae/drug effects , Cholesterol/chemistry , Endothelial Cells/drug effects , Liposomes/chemistry , Membrane Microdomains/drug effects , Transfection/methods , Animals , Caveolae/chemistry , Caveolae/metabolism , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line, Transformed , Cholesterol/metabolism , Clathrin/metabolism , DNA/chemistry , DNA/metabolism , Endocytosis/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Filipin/chemistry , Filipin/pharmacology , Gene Expression , Liposomes/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Nystatin/chemistry , Nystatin/pharmacology , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/pharmacology , Pinocytosis/drug effects , Plasmids/chemistry , Plasmids/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...