Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Biol Chem ; 299(6): 104774, 2023 06.
Article in English | MEDLINE | ID: mdl-37142218

ABSTRACT

Mitochondria are signaling organelles implicated in cancer, but the mechanisms are elusive. Here, we show that Parkin, an E3 ubiquitination (Ub) ligase altered in Parkinson's disease, forms a complex with the regulator of cell motility, Kindlin-2 (K2), at mitochondria of tumor cells. In turn, Parkin ubiquitinates Lys581 and Lys582 using Lys48 linkages, resulting in proteasomal degradation of K2 and shortened half-life from ∼5 h to ∼1.5 h. Loss of K2 inhibits focal adhesion turnover and ß1 integrin activation, impairs membrane lamellipodia size and frequency, and inhibits mitochondrial dynamics, altogether suppressing tumor cell-extracellular matrix interactions, migration, and invasion. Conversely, Parkin does not affect tumor cell proliferation, cell cycle transitions, or apoptosis. Expression of a Parkin Ub-resistant K2 Lys581Ala/Lys582Ala double mutant is sufficient to restore membrane lamellipodia dynamics, correct mitochondrial fusion/fission, and preserve single-cell migration and invasion. In a 3D model of mammary gland developmental morphogenesis, impaired K2 Ub drives multiple oncogenic traits of EMT, increased cell proliferation, reduced apoptosis, and disrupted basal-apical polarity. Therefore, deregulated K2 is a potent oncogene, and its Ub by Parkin enables mitochondria-associated metastasis suppression.


Subject(s)
Membrane Proteins , Ubiquitin-Protein Ligases , Cell Movement , Membrane Proteins/metabolism , Mitochondria/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Humans
2.
Small ; 20(15): e2306364, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37997202

ABSTRACT

Sonodynamic therapy (SDT) offers a remarkable non-invasive ultrasound (US) treatment by activating sonosensitizer and generating reactive oxygen species (ROS) to inhibit tumor growth. The development of multifunctional, biocompatible, and highly effective sonosensitizers remains a current priority for SDT. Herein, the first report that Mn(II) ions chelated Gd-TCPP (GMT) nanosheets (NSs) are synthesized via a simple reflux method and encapsulated with pluronic F-127 to form novel sonosensitizers (GMTF). The GMTF NSs produce a high yield of ROS under US irradiation due to the decreased highest occupied molecular orbital-lowest unoccupied molecular orbital gap energy (2.7-1.28 eV). Moreover, Mn(II) ions endow GMTF with a fascinating Fenton-like activity to produce hydroxyl radicals in support of chemodynamic therapy (CDT). It is also effectively used in magnetic resonance imaging (MRI) with high relaxation rate (r 1: 4.401 mM-1 s-1) to track the accumulation of NSs in tumors. In vivo results indicate that the SDT and CDT in combination with programmed cell death protein 1 antibody (anti-PD-1) show effective metastasis prevention effects, and 70% of the mice in the GMTF + US + anti-PD-1 group survived for 60 days. In conclusion, this study develops a sonosensitizer with promising potential for utilizing both MRI-guided SDT and CDT strategies.


Subject(s)
Colonic Neoplasms , Metal-Organic Frameworks , Neoplasms , Porphyrins , Ultrasonic Therapy , Animals , Mice , Reactive Oxygen Species , Magnetic Resonance Imaging , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/drug therapy , Porphyrins/pharmacology , Porphyrins/therapeutic use , Ions , Cell Line, Tumor
3.
Small ; 19(11): e2204747, 2023 03.
Article in English | MEDLINE | ID: mdl-36585358

ABSTRACT

As the foremost cause of cancer-related death, metastasis consists of three steps: invasion, circulation, and colonization. Only targeting one single phase of the metastasis cascade may be insufficient since there are many alternative routes for tumor cells to disseminate. Here, to target the whole cascade of metastasis, hybrid erythrocyte and tumor cell membrane-coated nanoparticle (Hyb-NP) is designed with dual functions of increasing circulation time and recognizing primary, circulating, and colonized tumors. After loading with monensin, a recently reported metastasis inhibitor, the delivery system profoundly reduces spontaneous metastasis in an orthotopic breast cancer model. Underlying mechanism studies reveal that Hyb-NP can deliver monensin to its action site in the Golgi apparatus, and in return, monensin can block the exocytosis of Hyb-NP from the Golgi apparatus, forming a reservoir-like subcellular structure. Notably, the Golgi apparatus reservoir displays three vital functions for suppressing metastasis initialization, including enhanced subcellular drug retention, metastasis-related cytokine release inhibition, and directional migration inhibition. Collectively, based on metastasis cascade targeting at the tissue level, further formation of the Golgi apparatus drug reservoir at the subcellular level provides a potential therapeutic strategy for cancer metastasis suppression.


Subject(s)
Monensin , Neoplasms , Humans , Monensin/pharmacology , Golgi Apparatus/ultrastructure , Cytoplasm
4.
Mol Pharm ; 18(7): 2622-2633, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34165313

ABSTRACT

Cancer metastasis is the major cause of cancer-related death; therefore, achieving suppression of tumor metastasis is a long-sought goal in cancer therapy. As the premetastatic niche acts as a prerequisite for tumor metastasis, it serves as an effective target for metastasis suppression. This study tests the feasibility of inhalable porous microspheres loaded with two premetastatic niche modulation agents, metformin and docosahexaenoic acid, as orthotopic delivery carriers for the reversion of lung premetastatic microenvironments and targeted suppression of tumor lung metastasis. The microspheres were prepared via an improved emulsion-solvent evaporation method and exhibit an excellent lung deposition, leading to significant inhibition of circulating tumor cells (CTCs)-endothelial cells adhesion, reduction of vascular permeability, and suppression of adhesion protein expression in lung premetastatic microenvironments. As a result, inhalable microspheres can prevent tumor lung metastasis formation excellently in vivo. Overall, this study proved that the encapsulation of metformin and docosahexaenoic acid in inhalable microspheres could be a promising strategy for tumor lung metastasis inhibition via orthotopically modulating premetastatic niche in the lungs.


Subject(s)
Breast Neoplasms/prevention & control , Docosahexaenoic Acids/chemistry , Hypoglycemic Agents/pharmacology , Lung Neoplasms/prevention & control , Metformin/pharmacology , Microspheres , Administration, Inhalation , Animals , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/chemistry , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Metformin/administration & dosage , Metformin/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Rats, Sprague-Dawley , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Nano Lett ; 19(6): 3548-3562, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31026397

ABSTRACT

Metastasis is the major cause of high mortality in cancer patients; thus, blocking the metastatic process is of critical importance for cancer treatments. The premetastatic niche, a specialized microenvironment with aberrant changes related to inflammation, allows the colonization of circulating tumor cells (CTCs) and serves as a potential target for metastasis prevention. However, little effort has been dedicated to developing nanomedicine to amend the premetastatic niche. Here this study reports a premetastatic niche-targeting micelle for the modulation of premetastatic microenvironments and suppression of tumor metastasis. The micelles are self-assembled with the oleate carbon chain derivative of metformin and docosahexaenoic acid, two anti-inflammatory agents with low toxicity, and coated with fucoidan for premetastatic niche-targeting. The obtained functionalized micelles (FucOMDs) exhibit an excellent blood circulation profile and premetastatic site-targeting efficiency, inhibit CTC adhesion to activated endothelial cells, alleviate lung vascular permeability, and reverse the aberrant expression of key marker proteins in premetastatic niches. As a result, FucOMDs prevent metastasis formation and efficiently suppress both primary-tumor growth and metastasis formation when combined with targeted chemotherapy. Collectively, the findings here provide proof of concept that the modulation of the premetastatic niche with targeted anti-inflammatory agents provides a potent platform and a safe and clinical translational option for the suppression of tumor metastasis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Docosahexaenoic Acids/administration & dosage , Metformin/administration & dosage , Neoplasm Metastasis/prevention & control , Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Animals , Anti-Inflammatory Agents/blood , Anti-Inflammatory Agents/therapeutic use , Docosahexaenoic Acids/blood , Docosahexaenoic Acids/therapeutic use , Lung/blood supply , Metformin/blood , Metformin/therapeutic use , Mice , Micelles , Neoplasm Metastasis/pathology , Neoplasms/pathology , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/pathology , Rats, Sprague-Dawley
6.
Cytotechnology ; 75(2): 103-113, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36969569

ABSTRACT

The expression spectra of connexin (Cx) isoforms were investigated in three mouse melanoma cell lines: B16-F1 (F1), B16-F10 (F10), and B16-BL6 (BL6). Metastatic potential intensity was higher in the order of F1, F10, and BL6. A remarkable behavior of Cx45 was found among 20 isoforms. The expression level of Cx45 was highest in F1 and lowest in BL6. It was inductively predicted that Cx45 might be a novel suppressor of metastasis. A Cx45-overexpressing BL6 cell line (Cx45 +BL6) was developed and its properties were compared with those of a wild-type cell line of BL6 (W-BL6). Compared to W-BL6, Cx45 +BL6 showed reduced wound healing, Transwell® permeability, and matrix metalloproteinase 9 expression, suggesting the suppression of cellular migration and invasion. The expression of E-cadherin and integrin ß1 in Cx45 +BL6 was also lower than in W-BL6, suggesting reduced cell adhesion. The decrease in cell adhesion was supported by the cell washing-out assay. In contrast, no difference between W-BL6 and Cx45 +BL6 was observed in cell proliferation, suggesting no effect on cell-cycle regulating factors. Finally, an in vivo assay revealed a significant decrease in the number of metastatic colonies of Cx45 +BL6 (176 ± 25/lung) in comparison with those of W-BL6 (252 ± 23/lung) in a mouse model. In conclusion, Cx45 is a novel suppressor of melanoma metastasis. Supplementary Information: The online version contains supplementary material available at 10.1007/s10616-022-00563-x.

7.
Exp Anim ; 72(2): 183-192, 2023 May 17.
Article in English | MEDLINE | ID: mdl-36288978

ABSTRACT

The metastasis of various cancers is promoted by hyperglycemia. In contrast, melanoma and colorectal cancer seemed to be exceptional. We confirmed that the metastasis of melanoma B16-F10 could be suppressed by hyperglycemia. It was attractive from the prognostic point of view of the prevention of metastasis, though the problem of the risk of diabetes remained. Then, the effect of moderate hyperglycemic condition was investigated using a pre-diabetic model mouse (GKKO mouse). The metastasis of B16-F10 cells to liver was focused and the number and volume of metastatic colonies in liver were analyzed. The medians of the number of metastatic colonies in GKKO mice were 0.57-fold (P=0.06) compared to control mice. Analysis of macrophage markers revealed upregulation of CD86, a tumor-suppressive M1-type marker, and downregulation of CD206, a tumor-promotive M2-type marker. A tendency of upregulation of Cxcl10, a pro-inflammatory cytokine was also observed. Regarding cellular activities of B16-F10, migration activity and invasion activity were reduced by moderate hyperglycemia. In conclusion, metastasis of B16-F10 cells to liver could be suppressed by moderate hyperglycemia without the risk of diabetes. This information should contribute to dietary planning during prognosis.


Subject(s)
Hyperglycemia , Melanoma , Animals , Mice , Melanoma/secondary , Liver , Mice, Inbred C57BL
8.
Radiother Oncol ; 175: 185-190, 2022 10.
Article in English | MEDLINE | ID: mdl-35537606

ABSTRACT

BACKGROUND AND PURPOSE: The FLASH effect is a potential breakthrough in radiotherapy because ultra-high dose-rate irradiation can substantially widen the therapeutic window. While the normal tissue sparing at high doses and short irradiation times has been demonstrated with electrons, photons, and protons, so far evidence with heavy ions is limited to in vitro cell experiments. Here we present the first in vivo results with high-energy 12C-ions delivered at an ultra-high dose rate. MATERIALS AND METHODS: LM8 osteosarcoma cells were subcutaneously injected in the posterior limb of female C3H/He mice 7 days before radiation exposure. Both hind limbs of the animals were irradiated with 240 MeV/n 12C-ions at ultra-high (18 Gy in 150 ms) or conventional dose rate (∼18 Gy/min). Tumor size was measured until 28 days post-exposure, when animals were sacrificed and lungs, limb muscles, and tumors were collected for further histological analysis. RESULTS: Irradiation with carbon ions was able to control the tumour both at conventional and ultra-high dose rate. FLASH decreases normal tissue toxicity as demonstrated by the reduced structural changes in muscle compared to conventional dose-rate irradiation. Carbon ion irradiation in FLASH conditions significantly reduced lung metastasis compared to conventional dose-rate irradiation and sham-irradiated animals. CONCLUSIONS: We demonstrated the FLASH effect in vivo with high-energy carbon ions. In addition to normal tissue sparing, we observed tumor control and a substantial reduction of lung metastasis in an osteosarcoma mouse model.


Subject(s)
Bone Neoplasms , Lung Neoplasms , Osteosarcoma , Female , Mice , Animals , Radiotherapy Dosage , Protons , Carbon/therapeutic use , Mice, Inbred C3H , Osteosarcoma/radiotherapy , Lung Neoplasms/radiotherapy , Bone Neoplasms/radiotherapy
9.
Pharmaceutics ; 13(11)2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34834295

ABSTRACT

Designing optimal (neo)adjuvant therapy is a crucial aspect of the treatment of non-small-cell lung carcinoma (NSCLC). Standard methods of chemotherapy, radiotherapy, and immunotherapy represent effective strategies for treatment. However, in some cases with high metastatic activity and high levels of circulating tumour cells (CTCs), the efficacy of standard treatment methods is insufficient and results in treatment failure and reduced patient survival. CTCs are seen not only as an isolated phenomenon but also a key inherent part of the formation of metastasis and a key factor in cancer death. This review discusses the impact of NSCLC therapy strategies based on a meta-analysis of clinical studies. In addition, possible therapeutic strategies for repression when standard methods fail, such as the administration of low-toxicity natural anticancer agents targeting these phenomena (curcumin and flavonoids), are also discussed. These strategies are presented in the context of key mechanisms of tumour biology with a strong influence on CTC spread and metastasis (mechanisms related to tumour-associated and -infiltrating cells, epithelial-mesenchymal transition, and migration of cancer cells).

10.
ACS Appl Mater Interfaces ; 13(26): 30350-30358, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34165951

ABSTRACT

The hepatocyte growth factor (HGF)/HGF receptor (Met) signaling pathway serves as a potential target for preventing tumor metastasis yet poorly explored. Here, we developed a Met-targeted nanoinhibitor to efficiently suppress metastasis via a multiple blockading HGF/Met signaling pathway. A biocompatible nanovector comprising multiple type of inhibitors enables interrupting extracellular domain dimerization and intracellular domain phosphorylation simultaneously. Such a comprehensive blockade of signaling pathway restrains unregulated tumor cell migration, invasion, and proliferation and thus remarkably suppresses metastasis in an orthotopic breast tumor model. This method provides a safe and effective option for metastasis inhibition via modulation of the cell signaling pathway. To our best knowledge, the strategy of the multiple blockading signaling pathway has not been reported for preventing tumor metastasis.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Lung Neoplasms/prevention & control , Nanoparticles/therapeutic use , Signal Transduction/drug effects , Animals , Antineoplastic Agents/chemistry , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/therapeutic use , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Crizotinib/chemistry , Crizotinib/therapeutic use , DNA/chemistry , DNA/therapeutic use , Female , Hepatocyte Growth Factor/metabolism , Indoles/chemistry , Lung Neoplasms/secondary , Mice, Inbred BALB C , Nanoparticles/chemistry , Polymers/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/metabolism
11.
Adv Sci (Weinh) ; 8(4): 2002834, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33643797

ABSTRACT

Mitochondria are highly involved in the metastasis of cancer cells. However, low permeability of mitochondria impedes the entry of anti-cancer drugs. Here, a self-assembled nanoparticle platform is designed that not only targets the DNA-intercalating agent doxorubicin to mitochondria but also enhances the specific penetration by opening the mitochondrial permeability transition pores (MPTPs). With drastic improvement in mitochondrial uptake, the drug delivery system results in substantial mitochondrial impairment leading to amplified induction of apoptosis, depletion of energy supply, and inhibition of numerous metastasis-associated proteins. As a consequence, the drug delivery system significantly inhibits the orthotopic tumor growth, and suppressed the metastasis of cancer cells detached from primary tumors. Additionally, the nanoparticle exhibits a potent effect on eradicating the metastasis of disseminated tumor cell from blood to lung. The results show that strategies of targeting mitochondria and unlocking MPTP are feasible and beneficial to mitigate both tumorigenesis and metastasis.

12.
Anticancer Res ; 40(2): 709-718, 2020 02.
Article in English | MEDLINE | ID: mdl-32014912

ABSTRACT

BACKGROUND/AIM: Prostate cancer (PCa) is the second most commonly diagnosed cancer in men. In contrast to localized disease, metastatic PCa leads to increased mortality. Kisspeptin (KISS1) functions as a metastasis suppressor in various cancers. The aim of this study was to detect the expression of KISS1 and its receptor GPR54 (KISS1R) in prostate cancer. MATERIALS AND METHODS: The expression of KISS1 and KISS1R was examined in prostate cancer tissue specimens after radical prostatectomy. RESULTS: A higher expression of KISS1 and KISS1R was shown in patients with localized tumors (Stage ≤IIb) compared to patients with advanced (Stage ≥III) tumor. High Gleason score PCa and higher prognostic groups patients showed a lower expression rate of both KISS1 and KISS1R. CONCLUSION: A down-regulation of KISS1-KISS1R system was detected in advanced prostate cancer. KISS1as tumor suppressor might be useful in the future for the diagnosis, risk assessment of prostate cancer progression, as well as a therapeutic target for aggressive tumors.


Subject(s)
Kisspeptins/biosynthesis , Prostatic Neoplasms/metabolism , Receptors, Kisspeptin-1/biosynthesis , Aged , Humans , Immunohistochemistry , Kisspeptins/metabolism , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Kisspeptin-1/genetics , Receptors, Kisspeptin-1/metabolism
13.
Adv Cancer Res ; 132: 111-37, 2016.
Article in English | MEDLINE | ID: mdl-27613131

ABSTRACT

Metastasis requires coordinated expression of multiple genetic cassettes, often via epigenetic regulation of gene transcription. BRMS1 blocks metastasis, but not orthotopic tumor growth in multiple tumor types, presumably via SIN3 chromatin remodeling complexes. Although there is an abundance of strong data supporting BRMS1 as a metastasis suppressor, the mechanistic data directly connecting molecular pathways with inhibition of particular steps in metastasis are not well defined. In this review, the data for BRMS1-mediated metastasis suppression in multiple tumor types are discussed along with the steps in metastasis that are inhibited.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Neoplasms/pathology , Repressor Proteins/metabolism , Humans , Neoplasm Metastasis
14.
Electron. j. biotechnol ; 53: 54-60, Sep.2021. ilus, tab, graf
Article in English | LILACS | ID: biblio-1451272

ABSTRACT

BACKGROUND Cancer is a life-threatening disease that affects approximately 18 million individuals worldwide. Breast cancer is the most common female neoplasm globally with more than 276,480 new cases of invasive breast cancer expected to be diagnosed in women in the U.S. alone in 2020. Genetic and epigenetic factors play role in the carcinogenesis and progression of this disease. In this study, MCF-7 adenocarcinoma cells were transfected with CRISPR/Cas9 plasmid to either knock out CDK11 or to activate CDH1. Treated cells were allografted into the mammary glands of female rats (150­190 g, 6­8 weeks) to evaluate the capability of these cells to control cancer progression and metastasis. RESULTS qPCR data revealed a significant downregulation of CDK11 and upregulation of CDH1. Cell cycle analysis and apoptosis assays indicated the knockout of CDK11 and simultaneous activation of CDH1 resulted in cell cycle arrest at G2/M phase and accumulation of cells at G2. Meanwhile, the percentage of cells that underwent late apoptosis increased in both genome editing hits. Histopathological sectioning data indicated that untransfected MCF-7 cells were capable of developing tumors in the mammary gland and initiation g angiogenesis. Transfected cells significantly restricted cancer cell infiltration/invasion by minimally localizing tumors and inhibiting angiogenesis. CONCLUSIONS Although further investigation is needed, the present data indicate the potentiality of using CRISPR/Cas9-based therapy as a promising approach to treat breast cancer. Impact: these data indicate targeting cancer-related genes via any genome editing tool might represent a novel approach to combat cancer.


Subject(s)
Animals , Female , Rats , Breast Neoplasms/genetics , Adenocarcinoma/genetics , Cdh1 Proteins/genetics , CRISPR-Associated Protein 9/genetics , Breast Neoplasms/secondary , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL