Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 11(15): e2304609, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38342629

ABSTRACT

Accumulating evidence suggests that changes in the tumor microenvironment caused by radiotherapy are closely related to the recurrence of glioma. However, the mechanisms by which such radiation-induced changes are involved in tumor regrowth have not yet been fully investigated. In the present study, how cranial irradiation-induced senescence in non-neoplastic brain cells contributes to glioma progression is explored. It is observed that senescent brain cells facilitated tumor regrowth by enhancing the peripheral recruitment of myeloid inflammatory cells in glioblastoma. Further, it is identified that astrocytes are one of the most susceptible senescent populations and that they promoted chemokine secretion in glioma cells via the senescence-associated secretory phenotype. By using senolytic agents after radiotherapy to eliminate these senescent cells substantially prolonged survival time in preclinical models. The findings suggest the tumor-promoting role of senescent astrocytes in the irradiated glioma microenvironment and emphasize the translational relevance of senolytic agents for enhancing the efficacy of radiotherapy in gliomas.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Glioblastoma/genetics , Astrocytes/pathology , Senotherapeutics , Brain Neoplasms/genetics , Cell Line, Tumor , Tumor Microenvironment
2.
J Exp Clin Cancer Res ; 43(1): 39, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38303029

ABSTRACT

BACKGROUND: Ubiquitination plays an important role in proliferating and invasive characteristic of glioblastoma (GBM), similar to many other cancers. Tripartite motif 25 (TRIM25) is a member of the TRIM family of proteins, which are involved in tumorigenesis through substrate ubiquitination. METHODS: Difference in TRIM25 expression levels between nonneoplastic brain tissue samples and primary glioma samples was demonstrated using publicly available glioblastoma database, immunohistochemistry, and western blotting. TRIM25 knockdown GBM cell lines (LN229 and U251) and patient derived GBM stem-like cells (GSCs) GBM#021 were used to investigate the function of TRIM25 in vivo and in vitro. Co-immunoprecipitation (Co-IP) and mass spectrometry analysis were performed to identify NONO as a protein that interacts with TRIM25. The molecular mechanisms underlying the promotion of GBM development by TRIM25 through NONO were investigated by RNA-seq and validated by qRT-PCR and western blotting. RESULTS: We observed upregulation of TRIM25 in GBM, correlating with enhanced glioblastoma cell growth and invasion, both in vitro and in vivo. Subsequently, we screened a panel of proteins interacting with TRIM25; mass spectrometry and co-immunoprecipitation revealed that NONO was a potential substrate of TRIM25. TRIM25 knockdown reduced the K63-linked ubiquitination of NONO, thereby suppressing the splicing function of NONO. Dysfunctional NONO resulted in the retention of the second intron in the pre-mRNA of PRMT1, inhibiting the activation of the PRMT1/c-MYC pathway. CONCLUSIONS: Our study demonstrates that TRIM25 promotes glioblastoma cell growth and invasion by regulating the PRMT1/c-MYC pathway through mediation of the splicing factor NONO. Targeting the E3 ligase activity of TRIM25 or the complex interactions between TRIM25 and NONO may prove beneficial in the treatment of GBM.


Subject(s)
Glioblastoma , Transcription Factors , Tripartite Motif Proteins , Humans , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/pathology , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , RNA Splicing Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
3.
Cancer Commun (Lond) ; 42(1): 17-36, 2022 01.
Article in English | MEDLINE | ID: mdl-34981670

ABSTRACT

BACKGROUND: Second-generation programmed cell death-protein 1/programmed death-ligand 1 (PD-1/PD-L1) inhibitors, such as bintrafusp alfa (M7824), SHR-1701, and YM101, have been developed to simultaneously block PD-1/PD-L1 and transforming growth factor-beta/transforming growth factor-beta receptor (TGF-ß/TGF-ßR). Consequently, it is necessary to identify predictive factors of lung cancer patients who are not only resistant to PD-1/PD-L1 inhibitors but also sensitive to bifunctional drugs. The purpose of this study was to search for such predictors. METHODS: Multivariable Cox regression was used to study the association between the clinical outcome of treatment with PD-1/PD-L1 inhibitors and lymphocyte recovery after lymphopenia in lung cancer patients. Murine CMT167 lung cancer cells were engineered to express the firefly luciferase gene and implanted orthotopically in the lung of syngeneic mice. Bioluminescence imaging, flow cytometry, and immunohistochemistry were employed to determine response to immunotherapy and function of tumor-infiltrating immune cells. RESULTS: For lung cancer patients treated with anti-PD-1/PD-L1 antibodies, poor lymphocyte recovery was associated with a shorter progression-free survival (PFS; P < 0.001), an accumulation of regulatory T cells (Tregs), and an elimination of CD8+ T cells in the peripheral blood. Levels of CD8+ T cells and Treg cells were also imbalanced in the tumors and peripheral immune organs of mice with poor lymphocyte recovery after chemotherapy. Moreover, these mice failed to respond to anti-PD-1 antibodies but remained sensitive to the anti-PD-L1/TGF-ßR fusion protein (SHR-1701). Consistently, SHR-1701 but not anti-PD-1 antibodies, markedly enhanced IFN-γ production and Ki-67 expression in peripheral CD8+ T cells from patients with impaired lymphocyte recovery. CONCLUSIONS: Lung cancer patients with poor lymphocyte recovery and suffering from persistent lymphopenia after previous chemotherapy are resistant to anti-PD-1/PD-L1 antibodies but might be sensitive to second-generation agents such as SHR-1701.


Subject(s)
B7-H1 Antigen , Lung Neoplasms , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes , Humans , Immune Checkpoint Inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics
4.
J Neurooncol ; 154(3): 275-283, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34476666

ABSTRACT

PURPOSE: Gliomas are the most common and aggressive malignant brain tumors and are associated with high mortality and incidence in humans. Despite rigorous multi-modal therapy, including surgery, chemotherapy and radiotherapy, patients with malignant glioma survive an average of 12-15 months following primary diagnosis. Therefore, new molecular biomarkers are urgently needed for diagnosis and targeted therapy. Here, we find that suppression of CKAP4 might inhibit glioma growth through regulation of Hippo signaling. METHODS: We examined the expression levels of CKAP4 through analysis of RNA sequencing data from GEPIA and CGGA databases. Then, Lentivirus was used to construct stable cell lines with knockout or overexpression of CKAP4. Next, the function of CKAP4 on glioma was investigated in vitro and in an orthotopic brain tumor model in mice. Lastly, luciferase reporter assay, immunofluorescence and immunoblotting were performed to explore the potential mechanism of how CKAP4 affects gliomas. RESULTS: CKAP4 is highly upregulated in glioma and high CKAP4 expressing tumors were associated with poor patient survival. And CKAP4 promotes malignant progression of gliomas via inhibiting Hippo signaling. CONCLUSION: CKAP4 has potential as a promising biomarker and can predict the prognosis of patients with gliomas. And targeting CKAP4 expression may be an effective therapeutic strategy for the treatment of human gliomas.


Subject(s)
Brain Neoplasms , Glioma , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Cytoskeleton/metabolism , Gene Expression Regulation, Neoplastic , Glioma/genetics , Hippo Signaling Pathway , Humans , Membrane Proteins/metabolism , Mice
5.
Cell Death Differ ; 28(1): 367-381, 2021 01.
Article in English | MEDLINE | ID: mdl-32814880

ABSTRACT

NF-κB signaling plays a critical role in tumor growth and treatment resistance in GBM as in many other cancers. However, the molecular mechanisms underlying high, constitutive NF-κB activity in GBM remains to be elucidated. Here, we screened a panel of tripartite motif (TRIM) family proteins and identified TRIM22 as a potential activator of NF-κB using an NF-κB driven luciferase reporter construct in GBM cell lines. Knockout of TRIM22 using Cas9-sgRNAs led to reduced GBM cell proliferation, while TRIM22 overexpression enhanced proliferation of cell populations, in vitro and in an orthotopic xenograft model. However, two TRIM22 mutants, one with a critical RING-finger domain deletion and the other with amino acid changes at two active sites of RING E3 ligase (C15/18A), were both unable to promote GBM cell proliferation over controls, thus implicating E3 ligase activity in the growth-promoting properties of TRIM22. Co-immunoprecipitations demonstrated that TRIM22 bound a negative regulator of NF-κB, NF-κB inhibitor alpha (IκBα), and accelerated its degradation by inducing K48-linked ubiquitination. TRIM22 also formed a complex with the NF-κB upstream regulator IKKγ and promoted K63-linked ubiquitination, which led to the phosphorylation of both IKKα/ß and IκBα. Expression of a non-phosphorylation mutant, srIκBα, inhibited the growth-promoting properties of TRIM22 in GBM cell lines. Finally, TRIM22 was increased in a cohort of primary GBM samples on a tissue microarray, and high expression of TRIM22 correlated with other clinical parameters associated with progressive gliomas, such as wild-type IDH1 status. In summary, our study revealed that TRIM22 activated NF-κB signaling through posttranslational modification of two critical regulators of NF-κB signaling in GBM cells.


Subject(s)
Glioblastoma/metabolism , Minor Histocompatibility Antigens/metabolism , NF-kappa B/metabolism , Repressor Proteins/metabolism , Tripartite Motif Proteins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Glioblastoma/pathology , Humans , I-kappa B Kinase/metabolism , Male , Mice , Mice, Nude , Minor Histocompatibility Antigens/genetics , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation , Repressor Proteins/genetics , Signal Transduction , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Xenograft Model Antitumor Assays
6.
Am J Cancer Res ; 10(2): 545-563, 2020.
Article in English | MEDLINE | ID: mdl-32195026

ABSTRACT

Brain metastasis is a major cause of mortality in melanoma patients. The blood-brain barrier (BBB) prevents most anti-tumor compounds from entering the brain, which significantly limits their use in the treatment of brain metastasis. One strategy in the development of new treatments is to assess the anti-tumor potential of drugs currently used in the clinic. Here, we tested the anti-tumor effect of the BBB-penetrating antipsychotic trifluoperazine (TFP) on metastatic melanoma. H1 and Melmet1 human metastatic melanoma cell lines were used in vitro and in vivo. TFP effects on viability and toxicity were evaluated in proliferation and colony formation assays. Preclinical, therapeutic efficacy was evaluated in NOD/SCID mice, after intracardial injection of tumor cells. Molecular studies using immunohistochemistry, western blots, immunofluorescence and transmission electron microscopy were used to gain mechanistic insight into the biological activity of TFP. Our results showed that TFP decreased cell viability and proliferation, colony formation and spheroid growth in vitro. The drug also decreased tumor burden in mouse brains and prolonged animal survival after injection of tumor cells (53.0 days vs 44.5 days), TFP treated vs untreated animals, respectively (P < 0.01). At the molecular level, TFP treatment led to increased levels of LC3B and p62 in vitro and in vivo, suggesting an inhibition of autophagic flux. A decrease in LysoTracker Red uptake after treatment indicated impaired acidification of lysosomes. TFP caused accumulation of electron dense vesicles, an indication of damaged lysosomes, and reduced the expression of cathepsin B, a main lysosomal protease. Acridine orange and galectin-3 immunofluorescence staining were evidence of TFP induction of lysosomal membrane permeabilization. Finally, TFP was cytotoxic to melanoma brain metastases based on the increased release of lactate dehydrogenase into media. Through knockdown experiments, the processes of TFP-induced lysosomal membrane permeabilization and cell death appeared to be STAT3 dependent. In conclusion, our work provides a strong rationale for further clinical investigation of TFP as an adjuvant therapy for melanoma patients with metastases to the brain.

7.
Oncogene ; 39(5): 1125-1139, 2020 01.
Article in English | MEDLINE | ID: mdl-31605013

ABSTRACT

The Hippo signaling pathway controls organ development and is also known, in cancer, to have a tumor suppressing role. Within the Hippo pathway, we here demonstrate, in human gliomas, a functional interaction of a transmembrane protein, prostate transmembrane protein, androgen induced 1 (PMEPA1) with large tumor suppressor kinase 1 (LATS1). We show that PMEPA1 is upregulated in primary human gliomas. The PMEPA1 isoform PMEPA1a was predominantly expressed in glioma specimens and cell lines, and ectopic expression of the protein promoted glioma growth and invasion in vitro and in an orthotopic xenograft model in nude mice. In co-immunoprecipitation experiments, PMEPA1a associated with the Hippo tumor suppressor kinase LATS1. This interaction led to a proteasomal degradation of LATS1 through recruitment of the ubiquitin ligase, neural precursor cell expressed, developmentally downregulated 4 (NEDD4), which led to silencing of Hippo signaling. Alanine substitution in PMEPA1a at PY motifs resulted in failed LATS1 degradation. Targeting of a downstream component in the Hippo signaling pathway, YAP, with shRNA, interfered with the growth promoting activities of PMEPA1a in vitro and in vivo. In conclusion, the presented work shows that PMEPA1a contributes to glioma progression by a dysregulation of the Hippo signaling pathway and thus represents a promising target for the treatment of gliomas.


Subject(s)
Disease Progression , Glioblastoma/pathology , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Signal Transduction , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Nedd4 Ubiquitin Protein Ligases/metabolism , Neoplasm Invasiveness , Proteasome Endopeptidase Complex/metabolism , Protein Isoforms/metabolism , Protein Stability
8.
Clin Cancer Res ; 25(22): 6868-6881, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31492748

ABSTRACT

PURPOSE: Long noncoding RNAs (lncRNA) have essential roles in diverse cellular processes, both in normal and diseased cell types, and thus have emerged as potential therapeutic targets. A specific member of this family, the SWI/SNF complex antagonist associated with prostate cancer 1 (SChLAP1), has been shown to promote aggressive prostate cancer growth by antagonizing the SWI/SNF complex and therefore serves as a biomarker for poor prognosis. Here, we investigated whether SChLAP1 plays a potential role in the development of human glioblastoma (GBM). EXPERIMENTAL DESIGN: RNA-ISH and IHC were performed on a tissue microarray to assess expression of SChLAP1 and associated proteins in human gliomas. Proteins complexed with SChLAP1 were identified using RNA pull-down and mass spectrometry. Lentiviral constructs were used for functional analysis in vitro and in vivo. RESULTS: SChLAP1 was increased in primary GBM samples and cell lines, and knockdown of the lncRNA suppressed growth. SChLAP1 was found to bind heterogeneous nuclear ribonucleoprotein L (HNRNPL), which stabilized the lncRNA and led to an enhanced interaction with the protein actinin alpha 4 (ACTN4). ACTN4 was also highly expressed in primary GBM samples and was associated with poorer overall survival in glioma patients. The SChLAP1-HNRNPL complex led to stabilization of ACTN4 through suppression of proteasomal degradation, which resulted in increased nuclear localization of the p65 subunit of NF-κB and activation of NF-κB signaling, a pathway associated with cancer development. CONCLUSIONS: Our results implicated SChLAP1 as a driver of GBM growth as well as a potential therapeutic target in treatment of the disease.


Subject(s)
Actinin/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , Ribonucleoproteins/metabolism , Signal Transduction , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Mice , Prognosis , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Transport , Proteolysis , RNA, Long Noncoding/chemistry , Ubiquitination , Xenograft Model Antitumor Assays
9.
Oncogene ; 38(37): 6414-6428, 2019 09.
Article in English | MEDLINE | ID: mdl-31332287

ABSTRACT

Increasing evidence demonstrates that ubiquitin specific protease 39 (USP39) plays an oncogenic role in various human tumors. Here, using expression analysis of the publicly available Oncomine database, clinical glioma patient samples, and glioma cells, we found that USP39 was overexpressed in human gliomas. Knockdown of USP39 in glioma cells demonstrated that the protein promoted cell growth, invasion and migration in vitro and in a tumor model in nude mice. To identify mediators of USP39 growth-promoting properties, we used luciferase reporter constructs under transcriptional control of various promoters specific to seven canonical cancer-associated pathways. Luciferase activity from a synthetic TEAD-dependent YAP/TAZ-responsive reporter, as a direct readout of the Hippo signaling pathway, was decreased by 92% in cells with USP39 knockdown, whereas the luciferase activities from the other six cancer pathways, including MAPK/ERK, MAPK/JNK, NFκB, Notch, TGFß, and Wnt, remained unchanged. TAZ protein expression however was decreased independent of canonical Hippo signaling. Immunohistochemistry revealed a positive correlation between USP39 and TAZ proteins in orthotopic xenografts derived from modified glioma cells expressing USP39 shRNAs and primary human glioma samples (p < 0.05). Finally, loss of USP39 decreased TAZ pre-mRNA splicing efficiency in glioma cells in vitro, which led to reduced levels of TAZ protein. In summary, USP39 has oncogenic properties that increase TAZ protein levels by inducing maturation of its mRNA. USP39 therefore provides a novel therapeutic target for the treatment of human glioma.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , RNA Splicing/genetics , Trans-Activators/genetics , Ubiquitin-Specific Proteases/physiology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/metabolism , Trans-Activators/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Ubiquitin-Specific Proteases/genetics
10.
Biomed Pharmacother ; 109: 39-46, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30391707

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive primary human brain tumor. Although comprehensive therapies combining radiotherapy and chemotherapy after surgery can prolong survival, the prognosis is still poor with a median survival of only 14.6 months. Chemoresistance is one of the major causes of relapse as well as poor survival in glioma patients. Therefore, novel strategies to overcome chemoresistance are desperately needed for improved treatment of human GBM. Recent studies have demonstrated that the tumor microenvironment plays a critical role in the chemoresistance of various tumor types, which makes it a suitable target in anti-cancer therapies, as well as a valuable biomarker for prognostic purposes. This review focuses on chemoresistance in GBM induced by stromal cells, including the endothelium of blood vessels, astrocytes, and myeloid cells, as well as non-cellular factors in the tumor microenvironment. Corresponding therapies are discussed, including progressive strategies involving 3-dimensional models integrating engineering as well as biological advances.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Animals , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Combined Modality Therapy , Drug Resistance, Neoplasm , Glioblastoma/pathology , Humans , Prognosis , Survival Rate , Tumor Microenvironment
11.
Cell Death Dis ; 9(5): 517, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29725063

ABSTRACT

Increased Actin-like 6A (ACTL6A) expression has been implicated in the development of diverse cancers and recently associated with the Hippo signaling pathway, which is known to regulate biological properties, including proliferation, tissue regeneration, stem cell biology, as well as tumorigenesis. Here we first show that ACTL6A is upregulated in human gliomas and its expression is associated with glioma patient survival. ACTL6A promotes malignant behaviors of glioma cells in vitro and in orthotopic xenograft model. In co-immunoprecipitation assays, we discover that ACTL6A physically associated with YAP/TAZ and furthermore disrupts the interaction between YAP and ß-TrCP E3 ubiquitin ligase, which promotes YAP protein degradation. Moreover, effects of ACTL6A on glioma cells proliferation, migration, and invasion could be mediated by YAP/TAZ. These data indicate that ACTL6A may contribute to cancer progression by stabilizing YAP/TAZ and therefore provide a novel therapeutic target for the treatment of human gliomas.


Subject(s)
Actins/genetics , Adaptor Proteins, Signal Transducing/genetics , Brain Neoplasms/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Intracellular Signaling Peptides and Proteins/genetics , Phosphoproteins/genetics , beta-Transducin Repeat-Containing Proteins/genetics , Actins/antagonists & inhibitors , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Aged , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Disease Progression , Female , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Phosphoproteins/metabolism , Protein Binding , Protein Stability , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Tumor Burden , Xenograft Model Antitumor Assays , YAP-Signaling Proteins , beta-Transducin Repeat-Containing Proteins/metabolism
12.
ACS Appl Mater Interfaces ; 10(1): 238-247, 2018 Jan 10.
Article in English | MEDLINE | ID: mdl-29243919

ABSTRACT

Porphyrin derivatives are the first-generation photosensitizers, and to design a strong near-infrared (NIR)-absorbing porphyrin with good water solubility is highly desired for better therapeutic effect to treat tumors. Herein, three new porphyrin derivatives, 5,10,15,20-tetrakis(3,4-dimethoxyphenyl) porphyrin (P1), 5,10,15,20-tetrakis(3,4-dimethoxyphenyl) zinc porphyrin (ZnP1), and 5,15-bis(3,4-dimethoxyphenyl)-10,20-bis((4-methoxyphenyl)ethynyl) zinc porphyrin (ZnP2) have been synthesized. Among them, ZnP2 shows the longest and most intensive Q-bands in the near-infrared (NIR) region, as it endows the strongest light-harvesting capability and deepest tumor tissue penetration. The three porphyrin derivatives were prepared into nanoparticles (NPs) via nanoprecipitation method, and the NPs exhibit good water dispersibility and passive tumor-targeting property through enhanced permeability and retention effect. Furthermore, these NPs demonstrate both photodynamic and photothermal effects. Through a systematic study of the singlet oxygen quantum yield and cytotoxicity of P1, ZnP1, and ZnP2 NPs in vitro on Hela cells, it is found that ZnP2 shows the highest singlet oxygen quantum yield (79%), and its NPs show the best therapeutic efficacy in vitro. In vivo experiments disclosed that ZnP2 NPs present high phototoxicity, low dark toxicity, and excellent biocompatibility, and could be used as promising photothermogenic photosensitizer for cancer treatment.


Subject(s)
Photosensitizing Agents/chemistry , HeLa Cells , Humans , Photochemotherapy , Porphyrins , Zinc
13.
ACS Appl Mater Interfaces ; 9(38): 32475-32481, 2017 Sep 27.
Article in English | MEDLINE | ID: mdl-28875695

ABSTRACT

Heavy atom effect and configuration are important for BODIPY derivatives to generate singlet oxygen (1O2) for photodynamic therapy. Herein, a series of BODIPY derivatives with different halogens were synthesized. 1O2 quantum yields (QYs) and MTT assay confirm that incorporation of more heavy atoms onto dimeric BODIPY cannot effectively enhance the 1O2 QYs. Rather, the dark toxicity increases. This phenomenon can be attributed to the competition of heavy atom effect and configuration of dimeric BODIPY. In addition the BODIPY derivative with two iodine atoms (BDPI) owns the highest 1O2 QYs (73%) and the lowest phototoxicity IC50 (1 µM). Furthermore, an in vivo study demonstrates that BDPI NPs can effectively inhibit tumor growth and can be used as a promising threanostic agent for photodynamic therapy in clinic.


Subject(s)
Boron Compounds/chemistry , Photochemotherapy , Singlet Oxygen
14.
J Mater Chem B ; 5(8): 1566-1573, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-32263929

ABSTRACT

Developing biocompatible, near infrared absorbing, and multi-functional photosensitizers is crucial for effective cancer phototherapy. In this contribution, a BF2 chelate of [4-iodo-5-(4-bromophenyl)-3-(4-methoxyphenyl)-1H-pyrrol-2-yl][4-iodo-5-(4-bromophenyl)-3-(4-methoxyphenyl)pyrrol-2-ylidene]amine (IABDP) with high singlet oxygen generation efficiency (∼92%) has been designed and synthesized. Soluble and near infrared absorbing nanoparticles (NPs) can be simply obtained from the self-assembly of IABDP molecules, which have a high photothermal conversion efficiency (∼37.9%). Under irradiation of a broadband Xenon lamp, IABBDP NPs are able to serve as common photosensitizers for photothermal imaging (PTI) and photoacoustic imaging (PAI) guided simultaneous photodynamic therapy (PDT) and photothermal therapy (PTT). Compared to the usual combined strategies that require two distinct photosensitizers and two excitation sources, the IABBDP NP based approach is simplified considerably, and hence is more convenient, reliable, and cost effective. Both in vitro and in vivo studies confirm the good biosafety and prominent anti-tumor phototoxicity of IABDP NPs. Finally, we demonstrate that the imaging guided synergistic dual modal phototherapy enabled by IABDP NPs can essentially inhibit tumor growth (87.2% inhibition) in mice without causing considerable side-effects, testifying the great potential of this multi-functional organic photosensitizer for clinical use.

15.
J Biomater Sci Polym Ed ; 27(6): 472-89, 2016.
Article in English | MEDLINE | ID: mdl-26764973

ABSTRACT

In this paper, a kind of glutathione-sensitive polymeric micelles was prepared through assembling in aqueous solution of an amphiphilic polymeric prodrug which was synthesized by linkage of 6-mercaptopurine (6-MP) and polyethylene glycol monomethyl ether using propiolic acid as a connecting arm. The glutathione (GSH)-sensitive strategy is based on a Michael addition-elimination reaction, that is the amphiphilic polymeric prodrug which contains α, ß-unsaturated carbonyl group acts as a Michael acceptor to receive the attack of nucleophile - glutathione, and undergoes elimination reaction to release the original drug. Transmission electron microscope observation showed that the polymeric micelles (PMs) had a spherical-like morphology with a mean diameter of 28 ± 3.2 nm. The dynamic light scattering investigation data exhibited that the size and distribution changes of PMs are negligible after being placed for 15 days. In vitro drug release study indicated that only less than 13% of 6-MP was released from the micelles under GSH stimulation at micromolar level, while 34.5, 53.7, and 77.8% accumulative release rates were achieved under GSH stimulation at millimolar level (1, 2 and 10 mM), respectively. The cell inhibition rate of PM solution against HL-60 cells carried out by MTT method reached 85%. The cellular uptake and the intracellular drug release of PMs in HL-60 cells were observed through determining the intracellular 6-MP content by UV-vis spectrophotometer. In vitro macrophage uptake study showed a low phagocytosis rate, indicating the long-circulation ability of the PMs.


Subject(s)
Drug Carriers/chemistry , Drug Liberation/drug effects , Glutathione/pharmacology , Micelles , Polyethylene Glycols/chemistry , Prodrugs/chemistry , Biological Transport/drug effects , Drug Carriers/metabolism , HL-60 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Intracellular Space/drug effects , Intracellular Space/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mercaptopurine/chemistry , Polyethylene Glycols/metabolism , Prodrugs/metabolism , Water/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...