Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 993
Filter
1.
Adv Sci (Weinh) ; 11(18): e2306950, 2024 May.
Article in English | MEDLINE | ID: mdl-38441365

ABSTRACT

Intracellular proteome aggregation is a ubiquitous disease hallmark with its composition associated with pathogenicity. Herein, this work reports on a cell-permeable photosensitizer (P8, Rose Bengal derivative) for selective photo induced proximity labeling and crosslinking of cellular aggregated proteome. Rose Bengal is identified out of common photosensitizer scaffolds for its unique intrinsic binding affinity to various protein aggregates driven by the hydrophobic effect. Further acetylation permeabilizes Rose Bengal to selectively image, label, and crosslink aggregated proteome in live stressed cells. A combination of photo-chemical, tandem mass spectrometry, and protein biochemistry characterizations reveals the complexity in photosensitizing pathways (both Type I & II), modification sites and labeling mechanisms. The diverse labeling sites and reaction types result in highly effective enrichment and identification of aggregated proteome. Finally, aggregated proteomics and interaction analyses thereby reveal extensive entangling of proteostasis network components mediated by HSP70 chaperone (HSPA1B) and active participation of autophagy pathway in combating proteasome inhibition. Overall, this work exemplifies the first photo induced proximity labeling and crosslinking method (namely AggID) to profile intracellular aggregated proteome and analyze its interactions.


Subject(s)
Photosensitizing Agents , Proteome , Photosensitizing Agents/metabolism , Proteome/metabolism , Humans , Rose Bengal/metabolism , Cross-Linking Reagents/metabolism , Proteomics/methods , Tandem Mass Spectrometry/methods , Protein Aggregates
2.
Int J Mol Sci ; 25(3)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38338976

ABSTRACT

This work was aimed at the complex analysis of the metabolic and oxygen statuses of tumors in vivo after photodynamic therapy (PDT). Studies were conducted on mouse tumor model using two types of photosensitizers-chlorin e6-based drug Photoditazine predominantly targeted to the vasculature and genetically encoded photosensitizer KillerRed targeted to the chromatin. Metabolism of tumor cells was assessed by the fluorescence lifetime of the metabolic redox-cofactor NAD(P)H, using fluorescence lifetime imaging. Oxygen content was assessed using phosphorescence lifetime macro-imaging with an oxygen-sensitive probe. For visualization of the perfused microvasculature, an optical coherence tomography-based angiography was used. It was found that PDT induces different alterations in cellular metabolism, depending on the degree of oxygen depletion. Moderate decrease in oxygen in the case of KillerRed was accompanied by an increase in the fraction of free NAD(P)H, an indicator of glycolytic switch, early after the treatment. Severe hypoxia after PDT with Photoditazine resulted from a vascular shutdown yielded in a persistent increase in protein-bound (mitochondrial) fraction of NAD(P)H. These findings improve our understanding of physiological mechanisms of PDT in cellular and vascular modes and can be useful to develop new approaches to monitoring its efficacy.


Subject(s)
NAD , Photochemotherapy , Animals , Mice , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/metabolism , Oxygen/metabolism , Disease Models, Animal , Photochemotherapy/methods
3.
ACS Appl Mater Interfaces ; 16(2): 2166-2179, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38170968

ABSTRACT

Hypoxia is a pervasive feature of solid tumors, which significantly limits the therapeutic effect of photodynamic therapy (PDT) and further influences the immunotherapy efficiency in breast cancer. However, the transient alleviation of tumor hypoxia fails to address the underlying issue of increased oxygen consumption, resulting from the rapid proliferation of tumor cells. At present, studies have found that the reduction of the oxygen consumption rate (OCR) by cytochrome C oxidase (COX) inhibition that induced oxidative phosphorylation (OXHPOS) suppression was able to solve the proposed problem. Herein, we developed a specific mitochondrial-targeting nanotrapper (I@MSN-Im-PEG), which exhibited good copper chelating ability to inhibit COX for reducing the OCR. The results proved that the nanotrapper significantly alleviated the hypoxic tumor microenvironment by copper chelation in mitochondria and enhanced the PDT effect in vitro and in vivo. Meanwhile, the nanotrapper improved photoimmunotherapy through both enhancing PDT-induced immunogenetic cell death (ICD) effects and reversing Treg-mediated immune suppression on 4T1 tumor-bearing mice. The mitochondrial-targeting nanotrapper provided a novel and efficacious strategy to enhance the PDT effect and amplify photoimmunotherapy in breast cancer.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Animals , Mice , Photochemotherapy/methods , Copper/pharmacology , Tumor Hypoxia , Cell Line, Tumor , Neoplasms/drug therapy , Hypoxia/drug therapy , Immunotherapy , Mitochondria/metabolism , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/metabolism , Tumor Microenvironment
4.
Adv Healthc Mater ; 13(4): e2302597, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37941492

ABSTRACT

Photodynamic therapy (PDT) is a promising approach for tumor treatment; however, the therapeutic resistance of cancer stem cells (CSCs) severely limits its efficacy and easily lead to recurrence. Herein, a hyaluronic acid (HA)-Ce6-Olaparib (OLA) micelle (HCCO) is developed, which combines the CSC targeting of HA, the PDT effect of Ce6, and the DNA damage repair inhibition of OLA. More importantly, HCCO induces immunogenic cell death (ICD) effects, promotes dendritic cells maturation, and alleviates myeloid-derived suppressor cells (MDSCs) infiltration to reverse CSC resistance. As a result, HCCO not only significantly inhibits the growth of 4T1 breast cancer cells and CSCs in vitro, but also effectively inhibits tumor recurrence and metastasis in vivo. This study provides a novel strategy for preventing tumor recurrence and metastasis by the combination of inhibiting DNA damage repair, reversing CSC resistance, and enhancing PDT.


Subject(s)
Hyaluronic Acid , Photochemotherapy , Humans , Hyaluronic Acid/pharmacology , Hyaluronic Acid/metabolism , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/prevention & control , Neoplastic Stem Cells/metabolism , Cell Line, Tumor , Photosensitizing Agents/metabolism
5.
Photochem Photobiol ; 100(1): 19-21, 2024.
Article in English | MEDLINE | ID: mdl-37212141

ABSTRACT

Log dose-response curves relating to the effect of mitochondrial photodamage on clonogenic survival using benzoporphyrin derivative. With wild-type cells, autophagy produces a "shoulder" on the curve which is absent when an ATG5 knockdown is examined. Loss of ATG5 prevents the process of autophagy, which is seen to be cytoprotective.


Subject(s)
Apoptosis , Photosensitizing Agents , Photosensitizing Agents/pharmacology , Photosensitizing Agents/metabolism , Autophagy , Mitochondria , Lysosomes , Cell Line, Tumor
6.
Eur J Med Chem ; 264: 115985, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38016298

ABSTRACT

The potential use of Ru(II) complexes as photosensitizers (PSs) in photodynamic therapy (PDT) has gained significant attention. In comparison with fluorophores with aggregation-caused quenching (ACQ), fluorophores with aggregation-induced emission (AIE) characteristics exhibit sustained fluorescence and dispersibility in aqueous solutions. PSs with AIE characteristics have received much attention in recent years. Herein, we reported two novel biotin-conjugated Ru(II) polypyridyl complexes (Ru1 and Ru2) with AIE characteristics. When exposed to 460 nm (10 mW cm-2) light, Ru1 and Ru2 exhibited outstanding photostability and photocatalytic activity. Ru1 and Ru2 could efficiently generate singlet oxygen and induce pUC19 DNA photolysis when exposed to 460 nm light. Interestingly, both Ru1 and Ru2 also functioned as catalysts for NADH oxidation when exposed to 460 nm light. The presence of biotin fragments in Ru1 and Ru2 enhanced the specific uptake of these complexes by tumor cells. Both complexes showed minimal toxicity to selected cells in the dark. Nevertheless, the phototoxicity of both complexes significantly increased upon 460 nm light irradiation for 15 min. Further experiments revealed that Ru2 primarily accumulated in mitochondria and might bind to mitochondrial DNA. Under 460 nm light irradiation, Ru2 induced the generation of reactive oxygen species (ROS) and NADH depletion disrupting intracellular redox homeostasis in A549 cells, activating the mitochondrial apoptosis pathway resulting in up-regulation of apoptotic marker caspase-3, effectively damaged A549 cell DNA and arrested A549 cell cycle in the S phase. In vivo anti-tumor experiments were conducted to assess the effects of Ru2 on tumor growth in A549 tumor-bearing mice. The results showed that Ru2 effectively inhibited tumor growth under 460 nm light irradiation conditions. These findings indicate that Ru2 has great potential as a targeted photosensitizer for mitochondrial targeting imaging and photodynamic therapy of tumors.


Subject(s)
Coordination Complexes , Photochemotherapy , Ruthenium , Animals , Mice , Photosensitizing Agents/pharmacology , Photosensitizing Agents/metabolism , Biotin/pharmacology , Biotin/metabolism , NAD/metabolism , Photochemotherapy/methods , Mitochondria/metabolism , Oxidation-Reduction , DNA/metabolism , Coordination Complexes/pharmacology , Coordination Complexes/metabolism , Ruthenium/pharmacology
7.
ACS Appl Mater Interfaces ; 16(1): 245-260, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38113527

ABSTRACT

Bladder cancer is characterized by high rates of recurrence and multifocality. Immunogenic cell death (ICD) of cancer cells has emerged as a promising strategy to improve the immunogenicity of tumor cells for enhanced cancer immunotherapy. Although photosensitizer-based photodynamic therapy (PDT) has been validated as capable of inducing ICD in cancer cells, the photosensitizers with a sufficient ICD induction ability are still rare, and there have been few reports on the development of advanced photosensitizers to strongly evoke the ICD of bladder cancer cells for eliciting potent antitumor immune responses and eradicating bladder carcinoma in situ. In this work, we have synthesized a new kind of endoplasmic reticulum (ER)-targeting aggregation-induced emission (AIE) photosensitizer (named DPASCP-Tos), which could effectively anchor to the cellular ER and trigger focused reactive oxygen species (ROS) production within the ER, thereby boosting ICD in bladder cancer cells. Furthermore, we have demonstrated that bladder cancer cells killed by ER-targeted PDT could serve as a therapeutic cancer vaccine to elicit a strong antitumor immunity. Prophylactic vaccination of the bladder cancer cells killed by DPASCP-Tos under light irradiation promoted the maturation of dendritic cells (DCs) and the expansion of tumor antigen-specific CD8+ T cells in vivo and protected mice from subsequent in situ bladder tumor rechallenge and extended animal survival. In summary, the ER-targeted AIEgens developed here significantly amplified the ICD of bladder cells through focused ROS-based ER oxidative stress and transformed bladder cancer cells into the therapeutic vaccine to enhance immunogenicity against orthotopic bladder cancer, providing valuable insights for bladder carcinoma treatment.


Subject(s)
Carcinoma , Neoplasms , Photochemotherapy , Urinary Bladder Neoplasms , Animals , Mice , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/metabolism , Reactive Oxygen Species/metabolism , CD8-Positive T-Lymphocytes , Immunogenic Cell Death , Urinary Bladder , Cell Line, Tumor , Neoplasms/drug therapy , Urinary Bladder Neoplasms/drug therapy , Endoplasmic Reticulum/metabolism , Immunotherapy , Carcinoma/drug therapy
8.
ACS Appl Bio Mater ; 6(12): 5776-5788, 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38061031

ABSTRACT

Photodynamic therapy (PDT) has emerged as an efficient and noninvasive treatment approach utilizing laser-triggered photosensitizers for combating cancer. Within this rapidly advancing field, iridium-based photosensitizers with their dual functionality as both imaging probes and PDT agents exhibit a potential for precise and targeted therapeutic interventions. However, most reported classes of Ir(III)-based photosensitizers comprise mononuclear iridium(III), with very few examples of dinuclear systems. Exploring the full potential of iridium-based dinuclear systems for PDT applications remains a challenge. Herein, we report a dinuclear Ir(III) complex (IRDI) along with a structurally similar monomer complex (IRMO) having 2-(2,4-difluorophenyl)pyridine and 4'-methyl-2,2'-bipyridine ligands. The comparative investigation of the mononuclear and dinuclear Ir(III) complexes showed similar absorption profiles, but the dinuclear derivative IRDI exhibited a higher photoluminescence quantum yield (Φp) of 0.70 compared to that of IRMO (Φp = 0.47). Further, IRDI showed a higher singlet oxygen generation quantum yield (Φs) of 0.49 compared to IRMO (Φs = 0.28), signifying the enhanced potential of the dinuclear derivative for image-guided photodynamic therapy. In vitro assessments indicate that IRDI shows efficient cellular uptake and significant photocytotoxicity in the triple-negative breast cancer cell line MDA-MB-231. In addition, the presence of a dual positive charge on the dinuclear system facilitates the inherent mitochondria-targeting ability without the need for a specific targeting group. Subcellular singlet oxygen generation by IRDI was confirmed using Si-DMA, and light-activated cellular apoptosis via ROS-mediated PDT was verified through various live-dead assays performed in the presence and absence of the singlet oxygen scavenger NaN3. Further, the mechanism of cell death was elucidated by an annexin V-FITC/PI flow cytometric assay and by investigating the cytochrome c release from mitochondria using Western blot analysis. Thus, the dinuclear complex designed to enhance spin-orbit coupling with minimal excitonic coupling represents a promising strategy for efficient image-guided PDT using iridium complexes.


Subject(s)
Coordination Complexes , Photochemotherapy , Triple Negative Breast Neoplasms , Humans , Photosensitizing Agents/metabolism , Iridium/pharmacology , Iridium/metabolism , Singlet Oxygen/metabolism , Coordination Complexes/pharmacology , Coordination Complexes/metabolism , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Mitochondria/metabolism
9.
ACS Appl Mater Interfaces ; 15(30): 35884-35894, 2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37487181

ABSTRACT

The effect of photodynamic therapy (PDT) is severely limited by tumor hypoxia and the short half-life of reactive oxygen species (ROS). Herein, we constructed a near-infrared (NIR) light-regulated PDT nanoplatform (TPP-UCNPs@MOF-Pt) consisting of an upconversion nanoparticle (UCNP) core and porphyrin-based metal-organic framework (MOF) shell with platinum nanoparticles (PtNPs) and a mitochondria-targeting triphenylphosphine (TPP) group on the surface. TPP-UCNPs@MOF-Pt could effectively relieve the tumor hypoxia by converting intracellular H2O2 to oxygen (O2) and elevated the ROS level to enhance PDT efficacy under NIR light irradiation. In addition, the mitochondria-targeting TPP-UCNPs@MOF-Pt was localized on the mitochondria, leading to severe depolarization of the mitochondrial membrane and activation of the apoptotic pathway, further amplifying the therapeutic efficacy. In vitro and in vivo experiments demonstrated that the greatly enhanced photodynamic therapeutic efficacy of TPP-UCNPs@MOF-Pt was achieved by combining relief of tumor hypoxia with mitochondrial targeting and NIR activation. This study provides a promising strategy for construction of an MOF-based multifunctional nanoplatform to address the current limitations of PDT treatment for hypoxic tumors.


Subject(s)
Metal Nanoparticles , Metal-Organic Frameworks , Nanoparticles , Neoplasms , Photochemotherapy , Humans , Metal-Organic Frameworks/pharmacology , Metal-Organic Frameworks/metabolism , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/metabolism , Platinum , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Oxygen/metabolism , Mitochondria/metabolism , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/metabolism , Cell Line, Tumor
10.
J Photochem Photobiol B ; 245: 112733, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37311303

ABSTRACT

In cells that are exposed to terrestrial sunlight, the indole moiety in the side chain of tryptophan (Trp) can suffer photo/oxidative damage (POD) by reactive oxygen species (ROS) and/or ultraviolet light (UV-B). Trp is oxidized to produce N-formylkynurenine (NFK), a UV-A-responsive photosensitizer that further degenerates into photosensitizers capable of generating ROS through exposure to visible light. Thus, Trp-containing proteins function as both victims, and perpetrators, of POD if they are not rapidly replaced through protein turnover. The literature indicates that protein turnover and DNA repair occur poorly in chromosomal interiors. We contend, therefore, that basic chromosomal proteins (BCPs) that are enveloped by DNA should have evolved to lack Trp residues in their amino acid sequences, since these could otherwise function as 'Trojan horse-type' DNA-damaging agents. Our global analyses of protein sequences demonstrates that BCPs consistently lack Trp residues, although DNA-binding proteins in general do not display such a lack. We employ HU-B (a wild-type, Trp-lacking bacterial BCP) and HU-B F47W (a mutant, Trp-containing form of the same bacterial BCP) to demonstrate that the possession of Trp is deleterious to BCPs and associated chromosomal DNA. Basically, we show that UV-B and UV-A (a) cause no POD in HU-B, but cause extensive POD in HU-B F47W (in vitro), as well as (b) only nominal DNA damage in bacteria expressing HU-B, but extensive DNA damage in bacteria expressing F47W HU-B (in vivo). Our results suggest that Trp-lacking BCPs could have evolved to reduce scope for protein-facilitated, sunlight-mediated damage of DNA by UV-A and visible light, within chromosomal interiors that are poorly serviced by protein turnover and DNA repair machinery.


Subject(s)
Bacterial Proteins , Chromosomes , DNA Damage , Genome , Histones , Oxidative Stress , Sunlight , Tryptophan , Humans , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/radiation effects , Chromosomes/chemistry , Chromosomes/metabolism , Chromosomes/radiation effects , Chromosomes, Bacterial/chemistry , Chromosomes, Bacterial/metabolism , Chromosomes, Bacterial/radiation effects , Escherichia coli/genetics , Escherichia coli/radiation effects , Genome/genetics , Genome/radiation effects , Histones/chemistry , Histones/metabolism , Histones/radiation effects , Hydrogen-Ion Concentration , In Situ Nick-End Labeling , Integration Host Factors/chemistry , Oxidation-Reduction/radiation effects , Phenylalanine/genetics , Photosensitizing Agents/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/chemistry , Tryptophan/deficiency , Tryptophan/genetics , Tryptophan/metabolism , Ultraviolet Rays
11.
Biomaterials ; 295: 122034, 2023 04.
Article in English | MEDLINE | ID: mdl-36746049

ABSTRACT

Radioresistance of Cancer stem cell (CSC) is an important cause of tumor recurrence after radiotherapy (RT). Herein, we designed a type I aggregation-induced emission (AIE) photosensitiser-loaded biomimetic mesoporous organosilicon nanosystem (PMT) for precise depletion of CSC to prevent tumor recurrence after RT. This PMT system is composed of a type I AIE photosensitiser (TBP-2) loaded mesoporous organosilicon nanoparticles (MON) with an outer platelet membrane. The PMT system is able to specifically target CSC. Intracellular glutathione activity leads to MON degradation and the release of TBP-2. Type I photodynamic therapy is activated by exposure to white light, producing a large amount of hydroxyl radicals to promote CSC death. The results of in vivo experiments demonstrated specific removal of CSC following PMT treatment, with no tumor recurrence observed when combined with RT. However, tumor recurrence was observed in mice that received RT only. The expression of CSC markers was significantly reduced following PMT treatment. We demonstrate the development of a system for the precise removal of CSC with good biosafety and high potential for clinical translation. We believe the PMT nanosystem represents a novel idea in the prevention of tumor recurrence.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Animals , Mice , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/metabolism , Biomimetics , Neoplastic Stem Cells/pathology , Photochemotherapy/methods , Cell Line, Tumor , Neoplasms/drug therapy
12.
J Photochem Photobiol B ; 239: 112645, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36608400

ABSTRACT

Photodynamic therapy (PDT) elicits cell death, vascular damage, or/and anti-tumor host immune response upon activating the administered photosensitive drug by an appropriate light source. Because PDT is heavily dependent on tissue oxygen (O2) in essence, the concentration-dependent impact of O2 on tailoring cellular response to PDT remains an in-depth investigation. As a multifaceted modality, optimal combinations of photosensitizer (PS) concentration, light dose, and O2 delivery are critical to achieve ideal therapeutic outcomes. We herein present a fully integrated all-in-one device for the in vitro assessment of PDT efficacy synchronizing the quantitative control of three PDT disciplines simultaneously, aiming at 1) identifying the influence of varying gaseous microenvironments on PDT; and 2) determining the contribution of each PDT factor and estimating the strength of their synergic effect. The gas-gradient-generating unit for contactless headspace O2 delivery and spatial light control filtering layer in our device could either work as a stand-alone module or combine to screen a range of experimental PDT parameters. By sweeping a total of 128 conditions over four 5-aminolevulinic acid (5-ALA) concentrations, four light dosages, and eight O2 levels in one single experiment, we determined the main effects of the three key PDT agents and highlighted the interactive effect between 5-ALA and light after full-factorial statistical analysis. Our device is not only a versatile tool for predicting PDT efficacy during the translational study but also provides valuable multidimensional information for the interrelation between key PDT factors, which may expedite clinical PDT dosimetry and furnish new insights for the fundamental understanding of photobiological processes.


Subject(s)
Photochemotherapy , Photochemotherapy/methods , Gases , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/metabolism , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Oxygen , Tumor Microenvironment
13.
Adv Mater ; 35(3): e2208555, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36255149

ABSTRACT

The success of tumor immunotherapy highlights the potential of harnessing immune system to fight cancer. Activating both native T cells and exhausted T cells is a critical step for generating effective antitumor immunity, which is determined based on the efficient presentation of tumor antigens and co-stimulatory signals by antigen-presenting cells, as well as immunosuppressive reversal. However, strategies for achieving an efficient antigen presentation process and improving the immunosuppressive microenvironment remain unresolved. Here, aggregation-induced-emission (AIE) photosensitizer-loaded nano-superartificial dendritic cells (saDC@Fs-NPs) are developed by coating superartificial dendritic cells membranes from genetically engineered 4T1 tumor cells onto nanoaggregates of AIE photosensitizers. The outer cell membranes of saDC@Fs-NPs are derived from recombinant lentivirus-infected 4T1 tumor cells in which peptide-major histocompatibility complex class I, CD86, and anti-LAG3 antibody are simultaneously anchored. These saDC@Fs-NPs could directly stimulate T-cell activation and reverse T-cell exhaustion for cancer immunotherapy. The inner AIE-active photosensitizers induce immunogenic cell death to activate dendritic cells and enhance T lymphocyte infiltration by photodynamic therapy, promoting the transformation of "cold tumors" into "hot tumors," which further boosts immunotherapy efficiency. This work presents a powerful photoactive and artificial antigen-presenting platform for activating both native T cells and exhausted T cells, as well as facilitating tumor photodynamic immunotherapy.


Subject(s)
Neoplasms , Photosensitizing Agents , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/metabolism , Antigens, Neoplasm , Immunotherapy , Immunosuppression Therapy , Neoplasms/therapy , Neoplasms/metabolism , Dendritic Cells , Cell Line, Tumor , Tumor Microenvironment
14.
Photochem Photobiol ; 99(2): 448-468, 2023 03.
Article in English | MEDLINE | ID: mdl-36117466

ABSTRACT

Ovarian cancer is the most lethal gynecologic malignancy with a stubborn mortality rate of ~65%. The persistent failure of multiline chemotherapy, and significant tumor heterogeneity, has made it challenging to improve outcomes. A target of increasing interest is the mitochondrion because of its essential role in critical cellular functions, and the significance of metabolic adaptation in chemoresistance. This review describes mitochondrial processes, including metabolic reprogramming, mitochondrial transfer and mitochondrial dynamics in ovarian cancer progression and chemoresistance. The effect of malignant ascites, or excess peritoneal fluid, on mitochondrial function is discussed. The role of photodynamic therapy (PDT) in overcoming mitochondria-mediated resistance is presented. PDT, a photochemistry-based modality, involves the light-based activation of a photosensitizer leading to the production of short-lived reactive molecular species and spatiotemporally confined photodamage to nearby organelles and biological targets. The consequential effects range from subcytotoxic priming of target cells for increased sensitivity to subsequent treatments, such as chemotherapy, to direct cell killing. This review discusses how PDT-based approaches can address key limitations of current treatments. Specifically, an overview of the mechanisms by which PDT alters mitochondrial function, and a summary of preclinical advancements and clinical PDT experience in ovarian cancer are provided.


Subject(s)
Ovarian Neoplasms , Photochemotherapy , Female , Humans , Drug Resistance, Neoplasm , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/metabolism , Ovarian Neoplasms/drug therapy , Mitochondria/metabolism , Cell Line, Tumor
15.
J Plast Surg Hand Surg ; 57(1-6): 324-329, 2023.
Article in English | MEDLINE | ID: mdl-35522455

ABSTRACT

Photodynamic therapy (PDT) is a new therapy for treating cancer with less toxicity, high selectivity, good cooperativity, and repetitive usability. However, keloid treatment by PDT is mainly focused on clinical appearance, and few studies have been conducted on the mechanisms of PDT. In this study, key factors of the classical mitochondrial apoptosis signaling pathway were measured to assess the effect of a new PDT photosensitizer (p1). A specific inhibitor of caspase-8 (Z-IETD-FMK) was also used to verify the possible mechanisms. Twelve samples were obtained from 12 patients (six with keloids and six without) selected randomly from the Department of Plastic Surgery at Peking Union Medical College Hospital from January to December 2020. After cell culture, fibroblasts were divided into 13 groups. The morphology of fibroblasts in each group was observed by microscopy. Cell activity was measured by cell counting kit-8, and cell apoptotic morphology was observed by TUNEL staining. The reactive oxygen species (ROS) relative value was measured by a ROS test kit. The expression levels of key mitochondrial factors (caspase-3, caspase-8, cytochrome-c, Bax, and Bcl-2) were assessed by western blot, and mRNA expression of caspase-3 and caspase-8 was measured by RT-qPCR. We showed that p1 had a satisfactory proapoptotic effect on keloid fibroblasts by increasing the expression of ROS, caspase-3, caspase-8, and cytochrome-c, and decreasing the Bcl-2/Bax ratio; however, this effect was partially inhibited by Z-IETD-FMK, indicating that caspase-8 may be one of the p1's targets to achieve the proapoptotic effect.


Subject(s)
Keloid , Photochemotherapy , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/metabolism , Photosensitizing Agents/therapeutic use , Caspase 3/metabolism , Caspase 3/pharmacology , Caspase 3/therapeutic use , Keloid/drug therapy , Keloid/pathology , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Reactive Oxygen Species/therapeutic use , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/pharmacology , Caspase 8/metabolism , Caspase 8/pharmacology , Caspase 8/therapeutic use , Apoptosis , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/pharmacology , Proto-Oncogene Proteins c-bcl-2/therapeutic use , Fibroblasts/pathology , Cytochromes/metabolism , Cytochromes/pharmacology , Cytochromes/therapeutic use
16.
Photochem Photobiol ; 99(3): 906-907, 2023.
Article in English | MEDLINE | ID: mdl-36039569

ABSTRACT

"Nobody expects the Spanish Inquisition." The color red has often been associated with danger and other adverse events. In the recent literature, the protein "KillerRed" has been proposed as a means for lethally sensitizing neoplastic cells and tissues to light. A somewhat different perspective on the feasibility of this approach is offered.


Subject(s)
Photochemotherapy , Green Fluorescent Proteins/metabolism , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/metabolism
17.
J Biophotonics ; 16(2): e202200160, 2023 02.
Article in English | MEDLINE | ID: mdl-36153307

ABSTRACT

Myopia and keratoconus have become common corneal diseases that threaten the quality of human vision, and keratoconus is one of the most common indications for corneal transplantation worldwide. Collagen crosslinking (CXL) using riboflavin and ultraviolet A (UVA) light is an effective approach for treating ophthalmic disorders and has been shown clinically not only to arrest further progression of keratoconus but also to improve refractive power for cornea. However, CXL surgery irradiated by UVA has various potential risks such as surface damage and endothelial cell damage. Here, near-infrared femtosecond laser-based two-photon CXL was first applied to ex vivo human corneal stroma, operating at low photon energy with high precision and stability. After two-photon CXL, the corneal stiffness can be enhanced by 300% without significantly reducing corneal transparency. These findings illustrate the optimized direction that depositing high pulses energy in corneal focal volume (not exceeding damage threshold), and pave the way to 3D CXL of in vivo human cornea with higher safety, precision, and efficacy.


Subject(s)
Keratoconus , Photochemotherapy , Humans , Keratoconus/drug therapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/metabolism , Cornea/metabolism , Corneal Stroma/metabolism , Ultraviolet Rays , Collagen/metabolism , Cross-Linking Reagents
18.
Front Biosci (Schol Ed) ; 15(4): 17, 2023 12 27.
Article in English | MEDLINE | ID: mdl-38163956

ABSTRACT

BACKGROUND: Mitochondrial dysfunction is considered an important mechanism in the pathogenesis of various diseases. Therefore, mitochondria are currently being considered as subjects for targeted therapies, particularly, phototherapy using 5-aminolevulinic acid. This study aimed to investigate the activity of mitochondria in cells with different mutation loads. MATERIALS AND METHODS: The study was conducted using 11 cybrid lines obtained from the THP-1 cell line (a human monocytic leukemia cell line) and platelets of patients with different mitochondrial mutations. RESULTS: Our results illustrate that 5-aminolevulinic acid was metabolized equally in all cell lines, however, there was a significant decrease in mitochondrial potential, which differed among lines. CONCLUSIONS: The results of this study can be used to develop a personalized therapeutic approach based on different mitochondrial activities.


Subject(s)
Aminolevulinic Acid , Photosensitizing Agents , Humans , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/metabolism , Photosensitizing Agents/pharmacology , Photosensitizing Agents/metabolism , Photosensitizing Agents/therapeutic use , Mitochondria/metabolism , Cell Line , THP-1 Cells , Cell Line, Tumor
19.
Curr Med Sci ; 42(6): 1267-1272, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36462133

ABSTRACT

OBJECTIVE: The aim of this study was to examine the effects of photodynamic therapy (PDT) on the expression of Nav1.7 in spinal dorsal root ganglion (DRG) neurons. METHODS: The primary DRG neurons from newborn SD rats were cultured. The cells were identified by neuron-specific enolase immunofluorescence staining. DRG neurons were divided into four groups: control group, photosensitizer group, laser group, and PDT group. The cell viability was detected by a cell counting kit-8 (CCK8) assay. qRT-PCR and Western blotting were used to determine the mRNA and protein expression levels of Nav1.7 in DRG neurons. RESULTS: The purity of the cultured primary DRG neurons was greater than 90%. Compared with the control group, no significant change was found in the cell viability of the photosensitizer group, while the viability in the laser group and the PDT group was significantly reduced. The mRNA and protein expression levels of Nav1.7 were significantly greater in the laser group and the PDT group than in the control group. At the same time, the mRNA and protein expression levels of Nav1.7 were greater in the laser group than in the PDT group. CONCLUSION: Both laser and PDT could upregulate the expression of Nav1.7 in DRG neurons, and the promoting effect might be related to the pain induced by clinical treatment. This study provides a research basis for the use of laser and PDT to treat pain. A better understanding of the relationship between Nav1.7 and PDT can help clinicians better manage PDT-related pain.


Subject(s)
Ganglia, Spinal , Photochemotherapy , Rats , Animals , Ganglia, Spinal/metabolism , Rats, Sprague-Dawley , Photosensitizing Agents/pharmacology , Photosensitizing Agents/metabolism , Pain , Neurons/metabolism , RNA, Messenger/metabolism
20.
J Biomed Opt ; 27(10)2022 10.
Article in English | MEDLINE | ID: mdl-36217225

ABSTRACT

Significance: Hypoxia imaging for surgical guidance has never been possible, yet it is well known that most tumors have microregional chronic and/or cycling hypoxia present as well as chaotic blood flow. The ability to image oxygen partial pressure (pO2) is therefore a unique control of tissue metabolism and can be used in a range of disease applications to understand the complex biochemistry of oxygen supply and consumption. Aim: Delayed fluorescence (DF) from the endogenous molecule protoporphyrin IX (PpIX) has been shown to be a truly unique reporter of the local oxygen partial pressure in tissue. PpIX is endogenously synthesized by mitochondria in most tissues, and the particular property of DF emission is directly related to low microenvironmental oxygen concentration. Here, it is shown that PpIX has a unique emission in hypoxic tumor tissue regions, which is measured as a DF signal in the red to near-infrared spectrum. Approach: A time-gated imaging system was used for PpIX DF for wide field direct mapping of pO2 changes. Acquiring both prompt and DF in a rapid sequential cycle allowed for imaging oxygenation in a way that was insensitive to the PpIX concentration. By choosing adequate parameters, the video rate acquisition of pO2 images could be achieved, providing real-time tissue metabolic information. Results: In this report, we show the first demonstration of imaging hypoxia signals from PpIX in a pancreatic cancer model, exhibiting >5X contrast relative to surrounding normal oxygenated tissues. Additionally, tissue palpation amplifies the signal and provides intuitive temporal contrast based upon neoangiogenic blood flow differences. Conclusions: PpIX DF provides a mechanism for tumor contrast that could easily be translated to human use as an intrinsic contrast mechanism for oncologic surgical guidance.


Subject(s)
Neoplasms , Protoporphyrins , Aminolevulinic Acid/metabolism , Fluorescence , Humans , Hypoxia/diagnostic imaging , Oxygen/metabolism , Photosensitizing Agents/metabolism , Protoporphyrins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...