Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Bioelectrochemistry ; 142: 107932, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34474205

ABSTRACT

Due to a lack of data on predictors of electroporation-based treatment outcomes, we investigated the potential predictive role of contrast-enhanced harmonic ultrasound (CEUS) in mice B16F10 melanoma treated by gene electrotransfer (GET) to silence melanoma cell adhesion molecule (MCAM) and radiotherapy, which has not been evaluated yet. CEUS evaluation was verified by tumor histological analysis. Mice bearing subcutaneous tumors were treated with GET to silence MCAM, irradiation or the combination of GET to silence MCAM and irradiation (combined treatment). CEUS of the tumors used to evaluate tumor perfusion was performed before and up to 10 days after the beginning of the experiment, and the CEUS results were compared with tumor growth and the number of blood vessels analyzed in the histological tumor sections. CEUS revealed a decrease in tumor perfusion in the combined therapy groups compared with the control groups and correlated with tumor histological analyses, which showed a decreased vascular density. In this study a trend of inverse correlation was observed between tumor perfusion and treatment efficacy. The greater the perfusion of the tumor, the shorter the expected doubling time. Furthermore, decreased perfusion showed a trend to correlate with higher antitumor efficacy. Thus, CEUS could be used to predict tumoral vascular density and treatment effectiveness.


Subject(s)
Electroporation/methods , Melanoma, Experimental , Ultrasonography/methods , Animals , Cell Line, Tumor , Female , Melanoma, Experimental/radiotherapy , Melanoma, Experimental/ultrastructure , Mice , Mice, Inbred C57BL
2.
Int J Biol Macromol ; 187: 481-491, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34298051

ABSTRACT

Heparan sulfate (HS) is involved in many biological activities, including the biogenesis and uptake of exosomes, which are related to the occurrence and development of tumors. This study investigated the role of HS analogues (heparin, low molecular weight heparin, and 6-O-desulfated heparin) in modulating exosome secretion, composition and functions. Exosomes derived from B16F10 cells exposed to different HS analogues were isolated and characterized by TEM, western blotting and Nanosight analyses. The number, size and protein cargo of exosomes secreted by HS analogues-induced B16F10 cells were detected. The findings indicated the reduced tumor-derived exosome secretion and protein cargo as reflected by lower levels of CD63, TSG101, heparinase and IL-6 in exosomes derived from heparin-induced B16F10 cells as compared with 6-O-desulfated heparin-induced tumor cells. Further functional assays demonstrated that exosomes from tumor cells exposed to heparin weakened tumor proliferation, migration and invasion most significantly among various exosomes derived from B16F10 cells treated with different HS analogues. Moreover, the sulfate group at 6-O position of heparan sulfate has been proved to play an important role in tumor-derived exosome formation and functions. This study suggested a vital view to develop more specific and efficient HS-based strategies in cancer treatment for targeting tumor-derived exosomes.


Subject(s)
Antineoplastic Agents/pharmacology , Exosomes/drug effects , Heparin, Low-Molecular-Weight/pharmacology , Heparin/analogs & derivatives , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA-Binding Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Exosomes/metabolism , Exosomes/ultrastructure , Heparin/pharmacology , Heparin Lyase/metabolism , Interleukin-6/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/ultrastructure , Mice , Neoplasm Invasiveness , Skin Neoplasms/metabolism , Skin Neoplasms/ultrastructure , Tetraspanin 30/metabolism , Transcription Factors/metabolism
3.
J Cell Biol ; 220(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34096975

ABSTRACT

How local interactions of actin regulators yield large-scale organization of cell shape and movement is not well understood. Here we investigate how the WAVE complex organizes sheet-like lamellipodia. Using super-resolution microscopy, we find that the WAVE complex forms actin-independent 230-nm-wide rings that localize to regions of saddle membrane curvature. This pattern of enrichment could explain several emergent cell behaviors, such as expanding and self-straightening lamellipodia and the ability of endothelial cells to recognize and seal transcellular holes. The WAVE complex recruits IRSp53 to sites of saddle curvature but does not depend on IRSp53 for its own localization. Although the WAVE complex stimulates actin nucleation via the Arp2/3 complex, sheet-like protrusions are still observed in ARP2-null, but not WAVE complex-null, cells. Therefore, the WAVE complex has additional roles in cell morphogenesis beyond Arp2/3 complex activation. Our work defines organizing principles of the WAVE complex lamellipodial template and suggests how feedback between cell shape and actin regulators instructs cell morphogenesis.


Subject(s)
Cell Membrane/metabolism , Cell Shape , Pseudopodia/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 2-3 Complex/metabolism , Animals , Cell Membrane/genetics , Cell Membrane/ultrastructure , Cell Movement , HEK293 Cells , HL-60 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Macrophages/metabolism , Macrophages/ultrastructure , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/ultrastructure , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Transport , Pseudopodia/genetics , Pseudopodia/ultrastructure , Signal Transduction , Time Factors , Wiskott-Aldrich Syndrome Protein Family/genetics
4.
Cells ; 9(4)2020 03 31.
Article in English | MEDLINE | ID: mdl-32244541

ABSTRACT

Melanoma is the most severe type of skin cancer. Its unique and heterogeneous metabolism, relying on both glycolysis and oxidative phosphorylation, allows it to adapt to disparate conditions. Mitochondrial function is strictly interconnected with mitochondrial dynamics and both are fundamental in tumour progression and metastasis. The malignant phenotype of melanoma is also regulated by the expression levels of the enzyme acid sphingomyelinase (A-SMase). By modulating at transcriptional level A-SMase in the melanoma cell line B16-F1 cells, we assessed the effect of enzyme downregulation on mitochondrial dynamics and function. Our results demonstrate that A-SMase influences mitochondrial morphology by affecting the expression of mitofusin 1 and OPA1. The enhanced expression of the two mitochondrial fusion proteins, observed when A-SMase is expressed at low levels, correlates with the increase of mitochondrial function via the stimulation of the genes PGC-1alpha and TFAM, two genes that preside over mitochondrial biogenesis. Thus, the reduction of A-SMase expression, observed in malignant melanomas, may determine their metastatic behaviour through the stimulation of mitochondrial fusion, activity and biogenesis, conferring a metabolic advantage to melanoma cells.


Subject(s)
Down-Regulation , Melanoma, Experimental/enzymology , Melanoma, Experimental/metabolism , Mitochondrial Dynamics , Sphingomyelin Phosphodiesterase/metabolism , Animals , Disease Models, Animal , Female , GTP Phosphohydrolases/metabolism , Melanoma, Experimental/ultrastructure , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/ultrastructure , Organelle Biogenesis , Oxidation-Reduction
5.
Int J Pharm ; 576: 119010, 2020 Feb 25.
Article in English | MEDLINE | ID: mdl-31901359

ABSTRACT

We previously reported that a weak current (WC, 0.3-0.5 mA/cm2) applied to cells can induce endocytosis to promote cytoplasmic delivery of hydrophilic macromolecules (MW: <70,000), such as dextran and siRNA, which leak from WC-induced endosomes into the cytoplasm (Hasan et al., 2016). In this study, we evaluated the characteristics of WC-mediated endocytosis for application of the technology to cytoplasmic delivery of macromolecular medicines. WC induced significantly higher cellular uptake of exogenous DNA fragments compared to untreated cells; the amount increased in a time-dependent manner, indicating that endocytosis was induced after WC. Moreover, following WC treatment of cells in the presence of an antibody (MW: 150,000) with the lysosomotropic agent chloroquine, the antibody was able to bind to its intracellular target. Thus, high molecular weight protein medicines delivered by WC-mediated endocytosis were functional in the cytoplasm. Transmission electron microscopy of cells treated by WC in the presence of gold nanoparticles covered with polyethylene glycol showed that the WC-induced endosomes exhibited an elliptical shape. In the WC-induced endosomes, ceramide, which makes pore structures in the membrane, was localized. Together, these results suggest that WC can induce unique endocytosis and that macromolecular medicines leak from endosomes through a ceramide pore.


Subject(s)
Antibodies, Monoclonal/metabolism , Cell Membrane/metabolism , DNA/metabolism , Endocytosis , Iontophoresis , Melanoma, Experimental/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Ceramides/metabolism , Cytoplasm/metabolism , DNA/administration & dosage , DNA/chemistry , Electric Conductivity , Endosomes/metabolism , Melanoma, Experimental/ultrastructure , Mice , Mice, Inbred BALB C , Molecular Weight , Porosity , Time Factors
6.
Sci Rep ; 9(1): 15329, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31653931

ABSTRACT

Exosomes are small extracellular vesicles (sEVs), playing a crucial role in the intercellular communication in physiological as well as pathological processes. Here, we aimed to study whether the melanoma-derived sEV-mediated communication could adapt to microenvironmental stresses. We compared B16F1 cell-derived sEVs released under normal and stress conditions, including cytostatic, heat and oxidative stress. The miRNome and proteome showed substantial differences across the sEV groups and bioinformatics analysis of the obtained data by the Ingenuity Pathway Analysis also revealed significant functional differences. The in silico predicted functional alterations of sEVs were validated by in vitro assays. For instance, melanoma-derived sEVs elicited by oxidative stress increased Ki-67 expression of mesenchymal stem cells (MSCs); cytostatic stress-resulted sEVs facilitated melanoma cell migration; all sEV groups supported microtissue generation of MSC-B16F1 co-cultures in a 3D tumour matrix model. Based on this study, we concluded that (i) molecular patterns of tumour-derived sEVs, dictated by the microenvironmental conditions, resulted in specific response patterns in the recipient cells; (ii) in silico analyses could be useful tools to predict different stress responses; (iii) alteration of the sEV-mediated communication of tumour cells might be a therapy-induced host response, with a potential influence on treatment efficacy.


Subject(s)
Extracellular Vesicles/metabolism , Melanoma, Experimental/pathology , Stress, Physiological , Animals , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Vesicles/drug effects , Extracellular Vesicles/ultrastructure , Male , Melanoma, Experimental/ultrastructure , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Proteome/metabolism , Silver/chemistry , Stress, Physiological/drug effects , Titanium/chemistry , Tumor Microenvironment/drug effects
7.
Cancer Lett ; 442: 396-408, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30439541

ABSTRACT

Intratumoral injection of chemotherapy agents may be employed in the treatment of cancers. However, its anti-tumor efficacy is significantly impeded by collagen fibers in the tumor which decrease drug penetration into the tumor tissues. To improve the penetration, collagen inhibiting drug exposure is required. In this study, microspheres were fabricated by the modified double emulsion-solvent evaporation method as the drug delivery system of losartan potassium (LP MSs), with 5% gelatin as the inner phase. The collagen inhibiting experiment analyzed by Sirius Red stains demonstrated that LP MSs may effectively inhibit collagen I synthesis in B16 tumors. In addition, 15% F127 was used as the solvent to fix the formulations at the injection site, with poly (α-l-glutamate) grafted polyethylene glycol mono methyl ether (PLG-g-mPEG)-cisplatin loaded nanoparticles (CDDP NPs) as the model drug. The in vivo live imaging system showed that formulations dissolved in 15% F127 had 54.91% CDDP NPs retained in tumors at the end of 10 days, in comparison with 19.72% for those solved in water, suggesting strong intratumoral retention property of the in situ gel. In addition, confocal laser scanning microscope (CLSM) and Energy-Dispersive Analysis of X-ray spectroscopy combined with scanning electron microscope (SEM-EDAX) tests showed that LP MSs can effectively enhance the distribution and penetration of CDDP NPs within tumors. Furthermore, tumors i.t. treated with LP MSs/CDDP NPs gel could be significantly halted, or even reduced to 200 mm3, comparing with a volume of about 12000 mm3 incontrol group at the end of the anti-tumor effect experiment. These results provided important guiding principles for prolonged and localized drug delivery system of intratumoral collagen inhibitor. The improvements of intratumoral penetration method made in this study provided practical significance for the treatment of cancer, especially for mass tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Drug Carriers , Fibrillar Collagens/biosynthesis , Gelatin/chemistry , Losartan/administration & dosage , Melanoma, Experimental/drug therapy , Polyethylene Glycols/chemistry , Polyglutamic Acid/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cisplatin/chemistry , Cisplatin/metabolism , Drug Compounding , Drug Liberation , Gels , Injections, Intralesional , Losartan/chemistry , Losartan/metabolism , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/ultrastructure , Mice, Inbred C57BL , Microspheres , Polyglutamic Acid/chemistry , Tissue Distribution , Tumor Burden/drug effects
8.
Angiogenesis ; 20(4): 599-613, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28875379

ABSTRACT

Blockade of the glycolytic activator PFKFB3 in cancer cells (using a maximum tolerable dose of 70 mg/kg of the PFKFB3 blocker 3PO) inhibits tumor growth in preclinical models and is currently being tested as a novel anticancer treatment in phase I clinical trials. However, a detailed preclinical analysis of the effects of such maximum tolerable dose of a PFKFB3 blocker on the tumor vasculature is lacking, even though tumor endothelial cells are hyper-glycolytic. We report here that a high dose of 3PO (70 mg/kg), which inhibits cancer cell proliferation and reduces primary tumor growth, causes tumor vessel disintegration, suppresses endothelial cell growth for protracted periods, (model-dependently) aggravates tumor hypoxia, and compromises vascular barrier integrity, thereby rendering tumor vessels more leaky and facilitating cancer cell intravasation and dissemination. These findings contrast to the effects of a low dose of 3PO (25 mg/kg), which induces tumor vessel normalization, characterized by vascular barrier tightening and maturation, but reduces cancer cell intravasation and metastasis. Our findings highlight the importance of adequately dosing a glycolytic inhibitor for anticancer treatment.


Subject(s)
Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Phosphofructokinase-2/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Melanoma, Experimental/ultrastructure , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Phosphofructokinase-2/metabolism , Pyridines/pharmacology
9.
Nat Med ; 23(7): 890-898, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28604702

ABSTRACT

The extracellular matrix (ECM) is a master regulator of cellular phenotype and behavior. It has a crucial role in both normal tissue homeostasis and disease pathology. Here we present a fast and efficient approach to enhance the study of ECM composition and structure. Termed in situ decellularization of tissues (ISDoT), it allows whole organs to be decellularized, leaving native ECM architecture intact. These three-dimensional decellularized tissues can be studied using high-resolution fluorescence and second harmonic imaging, and can be used for quantitative proteomic interrogation of the ECM. Our method is superior to other methods tested in its ability to preserve the structural integrity of the ECM, facilitate high-resolution imaging and quantitatively detect ECM proteins. In particular, we performed high-resolution sub-micron imaging of matrix topography in normal tissue and over the course of primary tumor development and progression to metastasis in mice, providing the first detailed imaging of the metastatic niche. These data show that cancer-driven ECM remodeling is organ specific, and that it is accompanied by comprehensive changes in ECM composition and topological structure. We also describe differing patterns of basement-membrane organization surrounding different types of blood vessels in healthy and diseased tissues. The ISDoT procedure allows for the study of native ECM structure under normal and pathological conditions in unprecedented detail.


Subject(s)
Basement Membrane/ultrastructure , Breast Neoplasms/ultrastructure , Extracellular Matrix/ultrastructure , Mammary Neoplasms, Experimental/ultrastructure , Proteomics , Tumor Microenvironment , Animals , Basement Membrane/metabolism , Breast Neoplasms/metabolism , Extracellular Matrix/metabolism , Female , Humans , Imaging, Three-Dimensional , Lactation , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lung Neoplasms/ultrastructure , Lymph Nodes/metabolism , Lymph Nodes/ultrastructure , Lymphatic Metastasis , Mammary Glands, Human/metabolism , Mammary Glands, Human/ultrastructure , Mammary Neoplasms, Experimental/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/ultrastructure , Mice , Optical Imaging , Peripheral Nerves/metabolism , Peripheral Nerves/ultrastructure , Tongue Neoplasms/metabolism , Tongue Neoplasms/ultrastructure
10.
Autophagy ; 13(8): 1331-1347, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28598240

ABSTRACT

Macroautophagy/autophagy is a dynamic and inducible catabolic process that responds to a variety of hormonal and environmental cues. Recent studies highlight the interplay of this central pathway in a variety of pathophysiological diseases. Although defective autophagy is implicated in melanocyte proliferation and pigmentary disorders, the mechanistic relationship between the 2 pathways has not been elucidated. In this study, we show that autophagic proteins LC3B and ATG4B mediate melanosome trafficking on cytoskeletal tracks. While studying melanogenesis, we observed spatial segregation of LC3B-labeled melanosomes with preferential absence at the dendritic ends of melanocytes. This LC3B labeling of melanosomes did not impact the steady-state levels of these organelles but instead facilitated their intracellular positioning. Melanosomes primarily traverse on microtubule and actin cytoskeletal tracks and our studies reveal that LC3B enables the assembly of microtubule translocon complex. At the microtubule-actin crossover junction, ATG4B detaches LC3B from melanosomal membranes by enzymatic delipidation. Further, by live-imaging we show that melanosomes transferred to keratinocytes lack melanocyte-specific LC3B. Our study thus elucidates a new role for autophagy proteins in directing melanosome movement and reveal the unconventional use of these proteins in cellular trafficking pathways. Such crosstalk between the central cellular function and housekeeping pathway may be a crucial mechanism to balance melanocyte bioenergetics and homeostasis.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy , Cysteine Endopeptidases/metabolism , Cytoskeleton/metabolism , Melanosomes/metabolism , Microtubule-Associated Proteins/metabolism , Movement , Actin Cytoskeleton/metabolism , Animals , Cytoskeleton/ultrastructure , Dendrites/metabolism , Gene Knockdown Techniques , Humans , Keratinocytes/metabolism , Lipids/chemistry , Melanocytes/metabolism , Melanocytes/ultrastructure , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/ultrastructure , Melanosomes/ultrastructure , Mice, Inbred C57BL , Microtubules/metabolism , Pigmentation
11.
Oncol Res ; 23(5): 229-36, 2016.
Article in English | MEDLINE | ID: mdl-27098146

ABSTRACT

RNA interference (RNAi) has been used for cancer gene therapy in recent years. However, the application of RNAi is hindered in the absence of safe and efficient gene delivery. In this article, a novel vehicle of graphene oxide functionalized with polyethylenimine and polyethylene glycol (GO-PEI-PEG) was successfully synthetized and then used to deliver plasmid-based Stat3 siRNA. The carrier can readily bind plasmid with high transfection efficiency. Moreover, molecular biology studies reveal that Stat3-related gene and protein expressions were significantly inhibited, suggesting that the formation of GO-PEI-PEG complexes could be utilized as a promising gene delivery in cancer therapy.


Subject(s)
Genetic Therapy/methods , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Plasmids/administration & dosage , RNA, Small Interfering/administration & dosage , STAT3 Transcription Factor/genetics , Transfection/methods , Animals , Cell Proliferation/genetics , Female , Gene Knockdown Techniques , Graphite/administration & dosage , Graphite/chemistry , Melanoma, Experimental/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Confocal/methods , Nanostructures/administration & dosage , Nanostructures/chemistry , Oxides/administration & dosage , Oxides/chemistry , Plasmids/genetics , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , RNA, Small Interfering/genetics , Random Allocation
12.
PLoS One ; 11(2): e0148980, 2016.
Article in English | MEDLINE | ID: mdl-26881822

ABSTRACT

In the present study we examined the ability of the amino acid derivative LTX-401 to induce cell death in cancer cell lines, as well as the capacity to induce regression in a murine melanoma model. Mode of action studies in vitro revealed lytic cell death and release of danger-associated molecular pattern molecules, preceded by massive cytoplasmic vacuolization and compromised lysosomes in treated cells. The use of a murine melanoma model demonstrated that the majority of animals treated with intratumoural injections of LTX-401 showed complete and long-lasting remission. Taken together, these results demonstrate the potential of LTX-401 as an immunotherapeutic agent for the treatment of solid tumors.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Antineoplastic Agents/pharmacology , Cytotoxins/pharmacology , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , beta-Alanine/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Antimicrobial Cationic Peptides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Cytotoxins/chemical synthesis , Erythrocytes/drug effects , Female , HMGB1 Protein/metabolism , Hemolysis/drug effects , Humans , Inhibitory Concentration 50 , Injections, Intralesional , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/ultrastructure , Melanoma, Experimental/metabolism , Melanoma, Experimental/ultrastructure , Mice , Mice, Inbred C57BL , Rats , Remission Induction , Skin Neoplasms/metabolism , Skin Neoplasms/ultrastructure , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/ultrastructure , beta-Alanine/chemical synthesis , beta-Alanine/pharmacology
13.
ACS Nano ; 10(1): 822-31, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26623481

ABSTRACT

Light-absorbing nanoparticles for localized heat generation in tissues have various biomedical applications in diagnostic imaging, surgery, and therapies. Although numerous plasmonic and carbon-based nanoparticles with strong optical absorption have been developed, their clearance, potential cytotoxicity, and long-term safety issues remain unresolved. Here, we show that "generally regarded as safe (GRAS)" melanoidins prepared from glucose and amino acid offer a high light-to-heat conversion efficiency, biocompatibility, biodegradability, nonmutagenicity, and efficient renal clearance, as well as a low cost for synthesis. We exhibit a wide range of biomedical photonic applications of melanoidins, including in vivo photoacoustic mapping of sentinel lymph nodes, photoacoustic tracking of gastrointestinal tracts, photothermal cancer therapy, and photothermal lipolysis. The biodegradation rate and renal clearance of melanoidins are controllable by design. Our results confirm the feasibility of biodegradable melanoidins for various photonic applications to theranostic nanomedicines.


Subject(s)
Biocompatible Materials/pharmacology , Melanoma, Experimental/therapy , Metal Nanoparticles/chemistry , Photons , Polymers/pharmacology , Theranostic Nanomedicine/methods , Amino Acids/chemistry , Animals , Biocompatible Materials/chemical synthesis , Biocompatible Materials/pharmacokinetics , Diagnostic Imaging/methods , Female , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/ultrastructure , Glomerular Filtration Rate , Glucose/chemistry , Gold/chemistry , Lipolysis/drug effects , Lymph Nodes/metabolism , Lymph Nodes/ultrastructure , Melanoma, Experimental/pathology , Melanoma, Experimental/ultrastructure , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Photoacoustic Techniques/methods , Phototherapy/methods , Polymers/chemical synthesis , Polymers/pharmacokinetics , Rats , Rats, Sprague-Dawley
14.
J Dermatol Sci ; 78(2): 133-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25818872

ABSTRACT

BACKGROUND: ADAMs (a disintegrin and metalloprotease) are a family of proteases involved in ectodomain shedding that play a role in various biological processes such as cell adhesion and migration. ADAM10 and ADAM17 are suggested to be involved in pigmentary disorders. OBJECTIVE: We examined the effect of ADAM protease inhibitors on the modulation of melanogenesis in normal human epidermal melanocytes (NHEM). METHODS: NHEMs and B16F10 treated with ADAM protease inhibitors were analyzed. AlamarBlue cell proliferation assay, melanin content assay, tyrosinase activity assay, Western blotting analysis, electron microscopic analysis, and RNA interference were employed. RESULTS: In NHEMs, melanin content was reduced by treatment with ADAM protease inhibitors. The inhibitors did not change the protein expression of tyrosinase, TRP-1, and MITF. In B16F10 cells, treatment of the cells with ADAM protease inhibitor diminished the α-MSH-induced increase in melanin content. Electron microscopy showed that the number of fibrillar and mature melanosomes was significantly reduced and that the vacuolar compartments were filled with dense unstructured aggregates after treatment with ADAM protease inhibitors. We therefore focused on the processing of PMEL17, a melanosomal glycoprotein that forms a fibrillar matrix on which melanin gets deposited. Proteolytic processing of PMEL17 is required to form functional fibrils during melanogenesis. Recently, γ-secretase and ß-site amyloid precursor protein-cleaving enzyme 2 (BACE2) were found to cleave PMEL17. We found that ADAM protease inhibitors exerted effects on the processing of C-terminal and N-terminal fragments of PMEL17. Using BACE2 siRNA and γ-secretase inhibitor, we showed that ADAM protease inhibitor affected PMEL17 processing in a γ-secretase and BACE2-independent mechanism. CONCLUSION: Several proteases, including ADAM proteases, can contribute to the formation of fibrils and their assembly into sheets in melanosomes.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Melanins/biosynthesis , Melanocytes/metabolism , Melanoma, Experimental/metabolism , Protease Inhibitors/pharmacology , gp100 Melanoma Antigen/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Cell Line, Tumor , Dipeptides/pharmacology , Humans , Interferon Type I/metabolism , Melanocytes/drug effects , Melanocytes/ultrastructure , Melanoma, Experimental/ultrastructure , Melanosomes/drug effects , Melanosomes/ultrastructure , Microphthalmia-Associated Transcription Factor/metabolism , Microscopy, Electron , Monophenol Monooxygenase/metabolism , Pregnancy Proteins/metabolism , RNA, Small Interfering/pharmacology , alpha-MSH/metabolism
15.
Cell Death Dis ; 4: e783, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23990023

ABSTRACT

Metal and its oxide nanoparticles show ideal pharmacological activity, especially in anti-tumor therapy. Our previous study demonstrated that cuprous oxide nanoparticles (CONPs) selectively induce apoptosis of tumor cells in vitro. To explore the anti-tumor properties of CONPs in vivo, we used the particles to treat mouse subcutaneous melanoma and metastatic lung tumors, based on B16-F10 mouse melanoma cells, by intratumoral and systemic injections, respectively. The results showed that CONPs significantly reduced the growth of melanoma, inhibited the metastasis of B16-F10 cells and increased the survival rate of tumor-bearing mice. Importantly, the results also indicated that CONPs were rapidly cleared from the organs and that these particles exhibited little systemic toxicity. Furthermore, we observed that CONPs targeted the mitochondria, which resulted in the release of cytochrome C from the mitochondria and the activation of caspase-3 and caspase-9 after the CONPs entered the cells. In conclusion, CONPs can induce the apoptosis of cancer cells through a mitochondrion-mediated apoptosis pathway, which raises the possibility that CONPs could be used to cure melanoma and other cancers.


Subject(s)
Copper/pharmacology , Melanoma, Experimental/pathology , Mitochondria/metabolism , Nanoparticles/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Copper/toxicity , HeLa Cells , Humans , Male , Melanoma, Experimental/ultrastructure , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/ultrastructure , Models, Biological , Nanoparticles/toxicity , Neoplasm Metastasis , Signal Transduction/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure , Subcutaneous Tissue/drug effects , Subcutaneous Tissue/pathology
16.
Exp Dermatol ; 20(11): 938-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21913996

ABSTRACT

Three major difficulties must be overcome to establish a quantitative method for melanosomal transfer analysis: (i) establishing a three-dimensional co-culture reassuring direct melanocyte to keratinocyte transfer, (ii) separation of melanocytes and keratinocytes following co-culture and (iii) melanosome quantification in each cell population. Melanocytes and keratinocytes are cultured on the opposite sides of the porous membrane of hanging cell inserts (1µm pores, 2×10(6) pores/cm(2) ). Cell separation is performed after 3days of co-culture by simple trypsinisation. Melanosome quantification in separated cell populations was accomplished by an ELISA-like method using gp-100 as the antigen. Melanocytes and keratinocytes come into 'direct' contact through the pores, and melanosomal transfer is accomplished without cell passage through the membrane. Cell separation by simple trypsinisation results in pure melanocyte and keratinocyte populations. Melanosome quantification by the ELISA-like method proved to be sensitive and specific to distinguish the known inhibitors and inducers of melanosomal transfer.


Subject(s)
Keratinocytes/ultrastructure , Melanocytes/ultrastructure , Melanosomes/ultrastructure , Spectrophotometry/methods , Animals , Cell Line, Tumor , Cell Separation , Coculture Techniques , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry , Melanoma, Experimental/ultrastructure , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission
17.
J Control Release ; 149(2): 190-5, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-20951750

ABSTRACT

Sonodynamic therapy (SDT) is a new modality using ultrasound (US) to activate certain chemical sensitizers for cancer therapy. In this study, the effect of US combined with a nanoparticle titanium dioxide (TiO(2)) on melanoma cell was investigated in vitro and in vivo. Melanoma cells (C32) were irradiated with US in the presence and/or absence of TiO(2). Cell viability was measured immediately after US irradiation (1MHz, 0.5 and 1.0W/cm(2) for 10s). The effect of the combination of TiO(2) and US exposure (1MHz, 1.0W/cm(2), 2 min duration) on subcutaneously implanted C32 solid tumors in mice were investigated by measuring tumor volume regression. The cell viability was significantly decreased only after US irradiation in the presence of TiO(2). In vivo results showed significant inhibition of tumor growth in groups treated with TiO(2) and US. To our knowledge, this is the first report to demonstrate the cell killing effect of TiO(2) nanoparticles under the irradiation US in vitro and in vivo.


Subject(s)
Melanoma, Experimental/therapy , Radiation-Sensitizing Agents/pharmacology , Titanium/pharmacology , Ultrasonic Therapy/methods , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Melanoma, Experimental/ultrastructure , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nanoparticles/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Radiation-Sensitizing Agents/radiation effects , Titanium/administration & dosage , Titanium/radiation effects , Xenograft Model Antitumor Assays
18.
Hum Gene Ther ; 22(3): 325-35, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21126174

ABSTRACT

Malignant melanoma is one of the leading causes of cancer mortality worldwide, underlining the need for effective novel therapies. In this study, the therapeutic efficacy and mechanism of systemic pro-opiomelanocortin (POMC) therapy were evaluated in mice bearing established melanoma. Injection of adenovirus encoding POMC (Ad-POMC) led to hepatic POMC overexpression and elevated adrenocorticotropin (ACTH) levels in the circulation. Systemic POMC therapy significantly attenuated the growth of established melanoma and prolonged the survival of tumor-bearing mice. Histological analysis revealed that systemic POMC therapy induced melanogenic differentiation while reducing melanoma growth. In addition, POMC therapy also elicited a significant reduction in the neovascular network of melanoma. Last, we demonstrated that POMC-derived peptides, including ACTH, α-melanocyte-stimulating hormone (α-MSH), and ß-MSH, are involved in POMC-mediated melanogenic differentiation and angiogenesis inhibition. In summary, systemic POMC therapy suppresses melanoma growth via induction of melanogenic differentiation and angiogenesis blockade, thereby demonstrating its potential as a novel treatment modality for melanoma.


Subject(s)
Cell Differentiation , Melanoma, Experimental/therapy , Neovascularization, Pathologic , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Adenoviridae/genetics , Animals , Cell Cycle/genetics , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Endothelial Cells/cytology , Genetic Therapy , Genetic Vectors/genetics , HEK293 Cells , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Melanoma, Experimental/ultrastructure , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/physiopathology , Survival Analysis , Xenograft Model Antitumor Assays
19.
Matrix Biol ; 29(6): 503-10, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20600893

ABSTRACT

Hyaluronan (HA) is a glycosaminoglycan composed of N-acetylglucosamine and glucuronic acid subunits. Endocytosis is thought to play an essential role in the catabolism of HA due to the intracellular compartmentalization of the HA degrading hyaluronidase enzymes. Previous investigations have shown that keratinocytes, chondrocytes and breast tumor cell lines endocytose HA via the cell surface glycoprotein, CD44. However, other cell types endocytose HA using a CD44-independent mechanism that remains to be defined. The purpose of this study was to investigate HA endocytosis in B16-F10 melanoma cells. We found that B16-F10 melanoma cells expressed CD44 on their surfaces. Unexpectedly, CD44 did not play a role in the endocytosis of HA. Electron microscopy studies revealed that B16-F10 melanoma cells exhibited membrane ruffling, a characteristic feature of macropinocytosis, only after incubating the cells with the HA co-polymer. Moreover, B16-F10 melanoma cells endocytosed HA via macropinocytosis as assessed by drug inhibition studies and the co-localization of fluorescently labeled HA with fluorescent tracers under confocal microscopy. Based on these results, we conclude that induced macropinocytosis may provide a previously unrecognized avenue for HA endocytosis in some cell types.


Subject(s)
Hyaluronic Acid/metabolism , Melanoma, Experimental/metabolism , Pinocytosis/physiology , Animals , Cell Line, Tumor , Chondrocytes/metabolism , Chondrocytes/pathology , Hyaluronoglucosaminidase/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/ultrastructure , Mice
20.
Lab Invest ; 90(6): 940-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20212453

ABSTRACT

This study analyzed whether therapy with CAMEL, an antimicrobial peptide (KWKLFKKIGAVLKVL), possess anticancer benefits. Although the peptide was cytotoxic for all the cell lines tested, it did not cause hemolysis, which suggests that CAMEL does not damage cell membranes. After cellular internalization, CAMEL localized to mitochondria and lowered the mitochondrial potential, resulting in the organelles' swelling, a decrease in cellular ATP level and, finally, cellular breakdown. High mobility group box 1 (HMGB1) protein, a necrotic death marker, was shown to be released from cells treated with CAMEL. Growth of B16-F10 melanoma tumors was clearly restrained after injections with CAMEL and could be kept in check throughout the period of peptide administration. However, if therapy was stopped, tumors started to grow again 3-4 days later. To reduce tumor volume and block tumor relapse, a combined therapy was required involving CAMEL and plasmid DNA carrying the interleukin-12 (IL-12) gene. The two therapeutic agents used in combination (a series of CAMEL injections first, followed by daily administration of plasmid DNA) delayed tumor growth and extended survival of treated animals in a statistically significant manner. Complete tumor regression was found in 60% of cases.


Subject(s)
Antineoplastic Agents/pharmacology , Oligopeptides/pharmacology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/ultrastructure , Cell Membrane/drug effects , Cell Membrane/physiology , Cell Survival/drug effects , Genetic Therapy/methods , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/ultrastructure , L-Lactate Dehydrogenase/metabolism , Melanoma/drug therapy , Melanoma/pathology , Melanoma/ultrastructure , Melanoma, Experimental/drug therapy , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Melanoma, Experimental/ultrastructure , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...