Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 293
Filter
1.
Methods Mol Biol ; 2839: 225-231, 2024.
Article in English | MEDLINE | ID: mdl-39008256

ABSTRACT

Radiolabeling enables the quantitation of newly synthesized heme and porphyrin, allowing us to distinguish heme synthesis rates from total cellular heme. Here, we describe a protocol for labeling heme with 14C-glycine or ALA and the sequential extraction of heme and porphyrin from the same samples for quantitation by liquid scintillation.


Subject(s)
Aminolevulinic Acid , Carbon Radioisotopes , Glycine , Heme , Porphyrins , Heme/chemistry , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/metabolism , Carbon Radioisotopes/chemistry , Porphyrins/chemistry , Glycine/chemistry , Isotope Labeling/methods , Humans
2.
Mol Pharm ; 21(7): 3218-3232, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38885477

ABSTRACT

Squamous cell carcinoma (SCC) is a common nonmelanoma skin cancer. Radiotherapy plays an integral role in treating SCC due to its characteristics, such as diminished intercellular adhesion, heightened cell migration and invasion capabilities, and immune evasion. These problems lead to inaccurate tumor boundary positioning and radiotherapy tolerance in SCC treatment. Thus, accurate localization and enhanced radiotherapy sensitivity are imperative for effective SCC treatment. To address the existing limitations in SCC therapy, we developed monoglyceride solid lipid nanoparticles (MG SLNs) and enveloped them with the A431 cell membrane (A431 CM) to create A431@MG. The characterization results showed that A431@MG was spherical. Furthermore, A431@MG had specific targeting for A431 cells. In A431 tumor-bearing mice, A431@MG demonstrated prolonged accumulation within tumors, ensuring precise boundary localization of SCC. We further advanced the approach by preparing MG SLNs encapsulating 5-aminolevulinic acid methyl ester (MLA) and desferrioxamine (DFO) with an A431 CM coating to yield A431@MG-MLA/DFO. Several studies have revealed that DFO effectively reduced iron content, impeding protoporphyrin IX (PpIX) biotransformation and promoting PpIX accumulation. Simultaneously, MLA was metabolized into PpIX upon cellular entry. During radiotherapy, the heightened PpIX levels enhanced reactive oxygen species (ROS) generation, inducing DNA and mitochondrial damage and leading to cell apoptosis. In A431 tumor-bearing mice, the A431@MG-MLA/DFO group exhibited notable radiotherapy sensitization, displaying superior tumor growth inhibition. Combining A431@MG-MLA/DFO with radiotherapy significantly improved anticancer efficacy, highlighting its potential to serve as an integrated diagnostic and therapeutic strategy for SCC.


Subject(s)
Carcinoma, Squamous Cell , Cell Membrane , Nanoparticles , Radiation-Sensitizing Agents , Skin Neoplasms , Animals , Mice , Nanoparticles/chemistry , Humans , Cell Line, Tumor , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/administration & dosage , Cell Membrane/metabolism , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/administration & dosage , Lipids/chemistry , Xenograft Model Antitumor Assays , Deferoxamine/chemistry , Deferoxamine/pharmacology , Mice, Nude , Female , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Liposomes
3.
Biosens Bioelectron ; 261: 116467, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38901392

ABSTRACT

Light therapy is an effective approach for the treatment of a variety of challenging dermatological conditions. In contrast to existing methods involving high doses and large areas of illumination, alternative strategies based on wearable designs that utilize a low light dose over an extended period provide a precise and convenient treatment. In this study, we present a battery-free, skin-integrated optoelectronic patch that incorporates a coil-powered circuit, an array of microscale violet and red light emitting diodes (LEDs), and polymer microneedles (MNs) loaded with 5-aminolevulinic acid (5-ALA). These polymer MNs, based on the biodegradable composite materials of polyvinyl alcohol (PVA) and hyaluronic acid (HA), serve as light waveguides for optical access and a medium for drug release into deeper skin layers. Unlike conventional clinical photomedical appliances with a rigid and fixed light source, this flexible design allows for a conformable light source that can be applied directly to the skin. In animal models with bacterial-infected wounds, the experimental group with the combination treatment of metronomic photodynamic and light therapies reduced 2.48 log10 CFU mL-1 in bactericidal level compared to the control group, indicating an effective anti-infective response. Furthermore, post-treatment analysis revealed the activation of proregenerative genes in monocyte and macrophage cell populations, suggesting enhanced tissue regeneration, neovascularization, and dermal recovery. Overall, this optoelectronic patch design broadens the scope for targeting deep skin lesions, and provides an alternative with the functionality of standard clinical light therapy methods.


Subject(s)
Photochemotherapy , Animals , Photochemotherapy/methods , Mice , Humans , Polyvinyl Alcohol/chemistry , Aminolevulinic Acid/therapeutic use , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/administration & dosage , Biosensing Techniques , Hyaluronic Acid/chemistry , Wound Infection/drug therapy , Wound Infection/microbiology , Wound Infection/therapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Skin/radiation effects , Skin/microbiology , Equipment Design
4.
Artif Cells Nanomed Biotechnol ; 52(1): 270-277, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38696132

ABSTRACT

Spherical gold/polyacrylic acid (Au/PAA) polymer-inorganic Janus nanoparticles (JNPs) with simultaneous therapeutic and targeting functions were fabricated. The obtained Au/PAA JNPs were further selectively functionalized with folic acid (FA) and thiol PEG amine (SH-PEG-NH2) on Au sides to provide superior biocompatibility and active targeting, while the other PAA sides were loaded with 5-aminolevulinic acid (5-ALA) to serve as a photosensitizer (PS) for photodynamic therapeutic (PDT) effects on MCF-7 cancer cells. The PS loading of 5-ALA was found to be 83% with an average hydrodynamic size and z-potential of 146 ± 0.8 nm and -6.40 mV respectively for FA-Au/PAA-ALA JNPs. The in vitro PDT study of the JNPs on MCF-7 breast cancer cells under 636 nm laser irradiation indicated the cell viability of 24.7% ± 0.5 for FA-Au/PAA-ALA JNPs at the IC50 value of 0.125 mM. In this regard, the actively targeted FA-Au/PAA-ALA JNPs treatment holds great potential for tumour therapy with high cancer cell-killing efficacy.


Subject(s)
Aminolevulinic Acid , Breast Neoplasms , Gold , Photochemotherapy , Photosensitizing Agents , Humans , MCF-7 Cells , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/pharmacology , Gold/chemistry , Gold/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Nanoparticles/chemistry , Acrylic Resins/chemistry , Female , Folic Acid/chemistry , Cell Survival/drug effects
5.
ACS Appl Bio Mater ; 7(5): 2899-2910, 2024 05 20.
Article in English | MEDLINE | ID: mdl-38607995

ABSTRACT

Acne vulgaris is one of the most prevalent skin disorders; it affects up to 85% of adolescents and often persists into adulthood. Topical 5-aminolevulinic acid (ALA)-based photodynamic therapy (PDT) provides an alternative treatment for acne; however, its efficacy is greatly undermined by the limited skin permeability of ALA. Herein, biocompatible ionic liquids (ILs) based on aliphatic acid/choline were employed to enhance the dermal delivery of ALA, thereby improving the efficacy of PDT. In addition to the one-step delivery of ALA by utilizing ILs as carriers, a two-step strategy of pretreating the skin with blank ILs, followed by the administration of free ALA, was employed to test the IL-facilitated dermal delivery of ALA in vitro. The cumulative permeation of ALA through the excised rat skin after IL pretreatment was significantly greater than that in the untreated group, the 20% dimethyl sulfoxide (DMSO) penetration enhancer group, and the one-step group. The penetration efficiency was influenced by formulation and treatment factors, including the type of IL, pretreatment duration, water content in the ILs, and concentration of ALA. In rats, IL pretreatment facilitated faster, greater, and deeper ALA-induced protoporphyrin IX (PpIX) accumulation. Moreover, the IL pretreatment regimen significantly improved the efficacy of ALA-based PDT against acne vulgaris in a rat ear model. The model IL choline citrate ([Ch]3[Cit]1) had a moderate effect on the skin barrier. Trans-epidermal water loss could be recovered 1 h after IL treatment, but no irritation to the rat skin was detected after 7 days of consecutive treatment. It was concluded that biocompatible IL pretreatment enhances the penetration of ALA and thus facilitates the transformation of PpIX and improves the efficacy of PDT against acne vulgaris.


Subject(s)
Acne Vulgaris , Aminolevulinic Acid , Ionic Liquids , Photochemotherapy , Photosensitizing Agents , Skin , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/chemistry , Animals , Ionic Liquids/chemistry , Ionic Liquids/pharmacology , Rats , Acne Vulgaris/drug therapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Skin/metabolism , Skin/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Materials Testing , Particle Size , Rats, Sprague-Dawley , Skin Absorption/drug effects , Male
6.
Bioorg Med Chem ; 105: 117726, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38626642

ABSTRACT

5-Aminolevulinic acid (ALA) and its derivatives, serving as the endogenous precursor of the photosensitizer (PS) protoporphyrin IX (PpIX), successfully applied in tumor imaging and photodynamic therapy (PDT). ALA and its derivatives have been used to treat actinic keratosis (AK), basal cell carcinoma (BCC), and improve the detection of superficial bladder cancer. However, the high hydrophilicity of ALA and the conversion of PpIX to heme have limited the accumulation of PpIX, hindering the efficiency and potential application of ALA-PDT. This study aims to evaluate the PDT activity of three rationally designed series of ALA-HPO prodrugs, which were based on enhancing the lipophilicity of the prodrugs and reducing the labile iron pool (LIP) through HPO iron chelators to promote PpIX accumulation. Twenty-four ALA-HPO conjugates, incorporating amide, amino acid, and ester linkages, were synthesized. Most of the conjugates, exhibited no dark-toxicity to cells, according to bioactivity evaluation. Ester conjugates 19a-g showed promoted phototoxicity when tested on tumor cell lines, and this increased phototoxicity was strongly correlated with elevated PpIX levels. Among them, conjugate 19c emerged as the most promising (HeLa, IC50 = 24.25 ± 1.43 µM; MCF-7, IC50 = 43.30 ± 1.76 µM; A375, IC50 = 28.03 ± 1.00 µM), displaying superior photodynamic anticancer activity to ALA (IC50 > 100 µM). At a concentration of 80 µM, the fluorescence intensity of PpIX induced by compound 19c in HeLa, MCF-7, and A375 cells was 18.9, 5.3, and 2.8 times higher, respectively, than that induced by ALA. In conclusion, cellular phototoxicity showed a strong correlation with intracellular PpIX fluorescence levels, indicating the potential application of ALA-HPO conjugates in ALA-PDT.


Subject(s)
Aminolevulinic Acid , Antineoplastic Agents , Drug Screening Assays, Antitumor , Photochemotherapy , Photosensitizing Agents , Humans , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Structure-Activity Relationship , Molecular Structure , Pyridones/pharmacology , Pyridones/chemistry , Pyridones/chemical synthesis , Cell Line, Tumor , Protoporphyrins/chemistry , Protoporphyrins/pharmacology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Cell Survival/drug effects , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/chemical synthesis
7.
Carbohydr Polym ; 336: 122114, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38670775

ABSTRACT

5-aminolevulinic acid (5-ALA) has been fully demonstrated as a biodegradable, without resistance, and pollution-free pesticide. However, the lack of targeting and the poor adhesion result in a low utilization rate, limiting its practical application. Herein, a dew-responsive polymer pro-pesticide Pec-hyd-ALA was successfully synthesized by grafting 5-ALA onto the pectin (PEC) backbone via acid-sensitive acylhydrazone bonds. When the pro-pesticide is exposed to acid dew on plant surfaces at night, 5-ALA is released and subsequently converted to photosensitize (Protoporphyrin IX, PpIX)in plant cells, leading to its accumulation and promoting photodynamic inactivation (PDI). An inverted fluorescence microscope has verified the accumulation of tetrapyrrole in plant cells. In addition, the highly bio-adhesive PEC backbone effectively improved the wetting and retention of 5-ALA on leaves. The pot experiment also demonstrated the system's control effect on barnyard grass. This work provides a promising approach to improving the herbicidal efficacy of 5-ALA.


Subject(s)
Aminolevulinic Acid , Herbicides , Pectins , Photosensitizing Agents , Pectins/chemistry , Herbicides/chemistry , Herbicides/pharmacology , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Protoporphyrins/chemistry , Protoporphyrins/pharmacology , Plant Leaves/chemistry , Wettability
8.
IEEE Trans Biomed Eng ; 71(1): 295-306, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37535482

ABSTRACT

Spectroscopy is a popular technique for identifying and quantifying fluorophores in fluorescent materials. However, quantifying the fluorophore of interest can be challenging when the material also contains other fluorophores (baseline), particularly if the emission spectrum of the baseline is not well-defined and overlaps with that of the fluorophore of interest. In this work, we propose a method that is free from any prior assumptions about the baseline by utilizing fluorescence signals at multiple excitation wavelengths. Despite the nonlinearity of the model, a closed-form expression of the least squares estimator is also derived. To evaluate our method, we consider the practical case of estimating the contributions of two forms of protoporphyrin IX (PpIX) in a fluorescence signal. This fluorophore of interest is commonly utilized in neuro-oncology operating rooms to distinguish the boundary between healthy and tumor tissue in a type of brain tumor known as glioma. Using a digital phantom calibrated with clinical and experimental data, we demonstrate that our method is more robust than current state-of-the-art methods for classifying pathological status, particularly when applied to images of simulated clinical gliomas. To account for the high variability in the baseline, we are examining various scenarios and their corresponding outcomes. In particular, it maintains the ability to distinguish between healthy and tumor tissue with an accuracy of up to 87%, while the ability of existing methods drops near 0%.


Subject(s)
Brain Neoplasms , Glioma , Humans , Aminolevulinic Acid/chemistry , Spectrometry, Fluorescence , Glioma/chemistry , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Fluorescent Dyes
9.
J Colloid Interface Sci ; 621: 180-194, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35461133

ABSTRACT

Chemodynamic therapy when combined with chemotherapy opens up a new avenue for treatment of cancer. However, its development is still restricted by low targeting, high dose and toxic side effects. Herein, rational designing and construction of a new multifunctional platform with the core-shell structure 5-ALA@UiO-66-NH-FAM@CP1 (ALA = 5-aminolevulinic acid, CP1 = zirconium-pemetrexed (Zr-MTA)) has been performed. In this platform, CP1 acting as a shell is encapsulated with the UiO-66-NH2 to engender a core-shell structure that promotes and achieves a high MTA loading rate through high affinity between MTA and unsaturated Zr site of UiO-66-NH2. The 5-ALA and 5-carboxyl fluorescein (5-FAM) was successfully loaded and covalently combined with UiO-66-NH2 due to its high porosity and presence of amino groups. The characterization results indicated that the loading rate of MTA (41.03 wt%) of platform is higher than the reported values. More importantly, the in vitro and in vivo results also demonstrated that it has a good folate targeting ability and realizes high efficient antitumor activity by chemotherapy combied with photodynamic therapy (PDT). This newly developed multifunctional platform could provide a new idea for designing and constructing the carrier with chemotherapy and PDT therapy.


Subject(s)
Metal-Organic Frameworks , Organometallic Compounds , Photochemotherapy , Aminolevulinic Acid/chemistry , Metal-Organic Frameworks/chemistry , Pemetrexed/pharmacology , Phthalic Acids
10.
Inorg Chem ; 61(6): 2846-2863, 2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35104130

ABSTRACT

Silver-indium-sulfide quantum dots (AIS QDs) have potential applications in many areas, including biomedicine. Their lack of regulated heavy metals, unlike many commercialized QDs, stands out as an advantage, but the necessity for alloyed or core-shell structures and related costly and sophisticated processes for the production of stable and high quantum yield aqueous AIS QDs are the current challenges. The present study demonstrates the one-step aqueous synthesis of simple AgInS2 QD compositions utilizing for the first time either a polyethyleneimine/2-mercaptopropionic acid (AIS-PEI/2MPA) mixture or only 2-mercaptopropionic acid (AIS-2MPA) as the stabilizing molecules, providing a AgInS2 portfolio consisting of cationic and anionic AIS QDs, respectively, and tuneable emission. Small AIS QDs with long-term stability and high quantum yields (19-23%) were achieved at a molar ratio of Ag/In/S 1/10/10 in water without any dopant or a semiconductor shell. The theranostic potential of these cationic and anionic AIS QDs was also evaluated in vitro. Non-toxic doses were determined, and fluorescence imaging potential was demonstrated. More importantly, these QDs were electrostatically loaded with zwitterionic 5-aminolevulinic acid (ALA) as a prodrug to enhance the tumor availability of ALA and to improve ALA-induced porphyrin photodynamic therapy (PDT). This is the first study investigating the influence of nanoparticle charge on ALA binding, release, and therapeutic efficacy. Surface charge was found to be more critical in cellular internalization and dark toxicity rather than drug loading and release. Both QDs provided enhanced ALA release at acidic pH but protected the prodrug at physiological pH, which is critical for tumor delivery of ALA, which suffers from low bioavailability. The PDT efficacy of the ALA-loaded AIS QDs was tested in 2D monolayers and 3D constructs of HT29 and SW480 human colon adenocarcinoma cancer cell lines. The incorporation of ALA delivery by the AIS QDs, which on their own do not cause phototoxicity, elicited significant cell death due to enhanced light-induced ROS generation and apoptotic/necrotic cell death, reducing the IC50 for ALA dramatically to about 0.1 and 0.01 mM in anionic and cationic AIS QDs, respectively. Combined with simple synthetic methods, the strong intracellular photoluminescence of AIS QDs, good biocompatibility of especially the anionic AIS QDs, and the ability to act as drug carriers for effective PDT signify that the AIS QDs, in particular AIS-2MPA, are highly promising theranostic QDs.


Subject(s)
Aminolevulinic Acid/pharmacology , Antineoplastic Agents/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Quantum Dots/chemistry , Aminolevulinic Acid/chemical synthesis , Aminolevulinic Acid/chemistry , Anions/chemical synthesis , Anions/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cations/chemical synthesis , Cations/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Indium/chemistry , Optical Imaging , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Silver/chemistry , Sulfides/chemistry , Tumor Cells, Cultured , Water/chemistry
11.
J Biol Chem ; 298(3): 101643, 2022 03.
Article in English | MEDLINE | ID: mdl-35093382

ABSTRACT

Heme is a critical biomolecule that is synthesized in vivo by several organisms such as plants, animals, and bacteria. Reflecting the importance of this molecule, defects in heme biosynthesis underlie several blood disorders in humans. Aminolevulinic acid synthase (ALAS) initiates heme biosynthesis in α-proteobacteria and nonplant eukaryotes. Debilitating and painful diseases such as X-linked sideroblastic anemia and X-linked protoporphyria can result from one of more than 91 genetic mutations in the human erythroid-specific enzyme ALAS2. This review will focus on recent structure-based insights into human ALAS2 function in health and how it dysfunctions in disease. We will also discuss how certain genetic mutations potentially result in disease-causing structural perturbations. Furthermore, we use thermodynamic and structural information to hypothesize how the mutations affect the human ALAS2 structure and categorize some of the unique human ALAS2 mutations that do not respond to typical treatments, that have paradoxical in vitro activity, or that are highly intolerable to changes. Finally, we will examine where future structure-based insights into the family of ALA synthases are needed to develop additional enzyme therapeutics.


Subject(s)
5-Aminolevulinate Synthetase , Anemia, Sideroblastic , Genetic Diseases, X-Linked , 5-Aminolevulinate Synthetase/chemistry , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/metabolism , Anemia, Sideroblastic/enzymology , Anemia, Sideroblastic/genetics , Animals , Heme , Humans , Structure-Activity Relationship
12.
J Photochem Photobiol B ; 223: 112286, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34416476

ABSTRACT

Plasmodium falciparum, the causative organism of Malaria is a mosquito-borne parasitic disease which infects red blood cells (RBCs), where it multiplies rapidly and goes through different stages of its life cycle. When the parasite load exceeds >3% in the blood, malaria transforms into severe malaria which requires immediate attention as death occurs within hours to days. The increase in people traveling to malaria-endemic areas and resistance/partial resistance to most known antimalarial drugs has put the current management scheme in jeopardy. To improve the patient outcome at this point, the physician may opt to perform exchange transfusions from another individual as an adjunct therapy to reduce parasitized RBCs, but the strategy has many drawbacks, including chances of infection. These limitations can be mitigated if the patient's own blood is withdrawn/extracted, sterilized from the parasitic load and then re-transfused almost similar to what is done in extracorporeal blood treatment for sepsis, poisoning and graft versus host disease. Thus, in the present study a light-based photochemical approach, Photodynamic Therapy (PDT) built on delta-aminolevulinic acid-protoporphyrin IX (ALA-PpIX) synthesis is exploited. This modality was effective at destruction of both resistant and susceptible strains of parasites, including at a high load mimicking severe drug resistant malaria. The current findings have set the stage for concept of an ALA-PpIX based PDT platform, "the REAP (Rapid Elimination of Active Plasmodium) strategy". This approach provides an additional tool towards the defense against multi-drug resistant severe malaria, and other intracellular blood pathogens, dependent on heme-synthesis.


Subject(s)
Antimalarials/pharmacology , Light , Malaria/pathology , Photosensitizing Agents/pharmacology , Plasmodium falciparum/drug effects , Aminolevulinic Acid/chemistry , Antimalarials/chemistry , Antimalarials/therapeutic use , Drug Resistance/drug effects , Erythrocytes/parasitology , Humans , Kinetics , Malaria/drug therapy , Malaria/parasitology , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Plasmodium falciparum/isolation & purification , Protoporphyrins/chemistry , Severity of Illness Index
13.
Sci Rep ; 11(1): 7283, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33790357

ABSTRACT

Hexaminolevulinate (HAL) induced Protoporphyrin IX (PpIX) fluorescence is commonly used to differentiate cancer cells from normal cells in vivo, as for instance in blue light cystoscopy for bladder cancer diagnosis. A detailed approach is here provided to use this diagnostic principle ex vivo in an immunosensor device, towards enabling non-invasive cancer diagnostic from body fluids, such as urine. Several factors susceptible to affect the applicability of HAL-assisted diagnosis in body fluids were tested. These included the cell viability and its impact on PpIX fluorescence, the storage condition and shelf life of HAL premix reagent, light exposure (360-450 nm wavelengths) and its corresponding effect on both intensity and bleaching of the PpIX fluorescence as a function of the microscopy imaging conditions. There was no significant decrease in the viability of bladder cancer cells after 6 h at 4 °C (student's t-test: p > 0.05). The cellular PpIX fluorescence decreased in a time-dependent manner when cancer cells were kept at 4 °C for extended period of time, though this didn't significantly reduce the fluorescence intensity contrast between cancer and non-cancer cells kept in the same condition for 6 h. HAL premix reagent kept in long term storage at 4 °C induced stronger PpIX fluorescence than reagent kept in the - 20 °C freezer. The PpIX fluorescence was negatively affected by repeated light exposure but increased with illumination intensity and exposure time. Though this applied to both healthy and cancer cell lines, and therefore did not statistically improved the differentiation between cell types. This study revealed important experimental settings that need to be carefully considered to benefit from the analytical potential of HAL induced fluorescence when used in technologies for the diagnosis of cancer from body fluids.


Subject(s)
Aminolevulinic Acid/analogs & derivatives , Biosensing Techniques/methods , Immunologic Tests/methods , Photosensitizing Agents/chemistry , Urinary Bladder Neoplasms/pathology , Aminolevulinic Acid/chemistry , Biosensing Techniques/standards , Cell Line, Tumor , Cells, Cultured , Humans , Immunologic Tests/standards , Liquid Biopsy/methods , Liquid Biopsy/standards , Microfluidics/methods , Microfluidics/standards , Protoporphyrins/metabolism , Sensitivity and Specificity , Urinary Bladder Neoplasms/urine , Urothelium/metabolism , Urothelium/pathology
14.
J Photochem Photobiol B ; 218: 112191, 2021 May.
Article in English | MEDLINE | ID: mdl-33862352

ABSTRACT

The effectiveness of the conventional chemotherapy for cancer are compromised as the cancer cells advances in their malignancy level as they acquired drug resistance. In this study, we aimed to evaluate the efficiency of aminolevulinic acid-photodynamic therapy (ALA-PDT) against cancer of various malignancy levels, indicated by the expression level of receptor associated nuclear factor-κB ligand (RANKL), through the expression levels of ALA uptake transporters. We established a malignancy model by gradually increasing the cell density of cancer cells. Western blotting was used to study the expression levels of RANKL, ALA uptake transporters and the cell density-dependent Yes-associated protein (YAP) under different cell densities. The amount of protoporphyrin (PpIX) produced and cell viability were then studied using high performance liquid chromatography (HPLC) and ALA-PDT assay. Our study showed that the amount of PpIX production doubled in high cell density/cancer malignancy cultures and the effectiveness of ALA-PDT when subjected to light irradiation at 635 nm are significantly at higher cancer malignancy. We observed that the expression levels of ALA uptake transporters and YAP correlated with higher cell density/cancer malignancy, suggesting a possible relationship among these three factors. These findings suggest that ALA-PDT is more effective in cancer cells of higher malignancy due to the upregulation of transporters involved in ALA uptake.


Subject(s)
Aminolevulinic Acid/chemistry , Antineoplastic Agents/chemistry , Membrane Transport Proteins/metabolism , Neoplasms/radiotherapy , Photosensitizing Agents/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Aminolevulinic Acid/pharmacology , Antineoplastic Agents/pharmacology , Biological Transport , Cell Count , Cell Line, Tumor , Cell Survival , Chromatography, High Pressure Liquid , Humans , Light , Photochemotherapy , Photosensitizing Agents/pharmacology , Protoporphyrins/chemistry , Transcription Factors/metabolism , YAP-Signaling Proteins
15.
Photochem Photobiol Sci ; 20(4): 489-499, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33825180

ABSTRACT

Photodynamic therapy (PDT) is an effective procedure for the treatment of lesions diseases based on the selectivity of a photosensitising compound with the ability to accumulate in the target cell. Atherosclerotic plaque is a suitable target for PDT because of the preferential accumulation of photosensitisers in atherosclerotic plaques. Dendrimers are hyperbranched polymers conjugated to drugs. The dendrimers of ALA hold ester bonds that inside the cells are cleaved and release ALA, yielding PpIX production. The dendrimer 6m-ALA was chosen to perform this study since in previous studies it induced the highest porphyrin macrophage: endothelial cell ratio (Rodriguez et al. in Photochem Photobiol Sci 14:1617-1627, 2015). We transformed Raw 264.7 macrophages to foam cells by exposure to oxidised LDLs, and we employed a co-culture model of HMEC-1 endothelial cells and foam cells to study the affinity of ALA dendrimers for the foam cells. In this work it was proposed an in vitro model of atheromatous plaque, the aim was to study the selectivity of an ALA dendrimer for the foam cells as compared to the endothelial cells in a co-culture system and the type of cell death triggered by the photodynamic treatment. The ALA dendrimer 6m-ALA showed selectivity PDT response for foam cells against endothelial cells. A light dose of 1 J/cm2 eliminate foam cells, whereas less than 50% of HMEC-1 is killed, and apoptosis cell death is involved in this process, and no necrosis is present. We propose the use of ALA dendrimers as pro-photosensitisers to be employed in photoangioplasty to aid in the treatment of obstructive cardiovascular diseases, and these molecules can also be employed as a theranostic agent.


Subject(s)
Aminolevulinic Acid/pharmacology , Apoptosis/drug effects , Foam Cells/drug effects , Macrophages/drug effects , Photosensitizing Agents/pharmacology , Aminolevulinic Acid/chemistry , Animals , Cell Line , Coculture Techniques , Foam Cells/physiology , Humans , Macrophages/physiology , Mice , Photosensitizing Agents/chemistry
16.
Int J Mol Sci ; 22(4)2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33671436

ABSTRACT

Polyamidoamine PAMAM dendrimer generation 3 (G3) was modified by attachment of biotin via amide bond and glucoheptoamidated by addition of α-D-glucoheptono-1,4-lacton to obtain a series of conjugates with a variable number of biotin residues. The composition of conjugates was determined by detailed 1-D and 2-D NMR spectroscopy to reveal the number of biotin residues, which were 1, 2, 4, 6, or 8, while the number of glucoheptoamide residues substituted most of the remaining primary amine groups of PAMAM G3. The conjugates were then used as host molecules to encapsulate the 5-aminolevulinic acid. The solubility of 5-aminolevulinic acid increased twice in the presence of the 5-mM guest in water. The interaction between host and guest was accompanied by deprotonation of the carboxylic group of 5-aminolevulinic acid and proton transfer into internal ternary nitrogen atoms of the guest as evidenced by a characteristic chemical shift of resonances in the 1H NMR spectrum of associates. The guest molecules were most likely encapsulated inside inner shell voids of the host. The number of guest molecules depended on the number of biotin residues of the host, which was 15 for non-biotin-containing glucoheptoamidated G3 down to 6 for glucoheptoamidated G3 with 8 biotin residues on the host surface. The encapsulates were not cytotoxic against Caco-2 cells up to 200-µM concentration in the dark. All encapsulates were able to deliver 5-aminolevulinic acid to cells but aqueous encapsulates were more active in this regard. Simultaneously, the reactive oxygen species were detected by staining with H2DCFDA in Caco-2 cells incubated with encapsulates. The amount of PpIX was sufficient for induction of reactive oxygen species upon 30-s illumination with a 655-nm laser beam.


Subject(s)
Amides/chemistry , Aminolevulinic Acid/pharmacology , Biotin/chemistry , Dendrimers/chemistry , Drug Delivery Systems , Polyamines/chemistry , Aminolevulinic Acid/chemistry , Caco-2 Cells , Cell Death/drug effects , Cell Survival/drug effects , Dendrimers/chemical synthesis , Fluorescence , Humans , Intracellular Space/metabolism , Polyamines/chemical synthesis , Proton Magnetic Resonance Spectroscopy , Protoporphyrins/pharmacology , Reactive Oxygen Species/metabolism
17.
ACS Appl Mater Interfaces ; 13(7): 7955-7965, 2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33565868

ABSTRACT

Photodynamic therapy (PDT) is a new therapeutic strategy for hypertrophic scars (HSs), and nanoethosomes (ES) have attracted considerable attention as an efficient transdermal delivery system for PDT of HSs (HS-PDT). However, the delivery of photosensitizers and the hypoxic microenvironment of HSs limit HS-PDT efficacy. Consequently, functional transdermal ES (A/A-ES) that are loaded with the photosensitizer, 5-aminolevulinic acid (ALA), and immobilized nanoenzyme Au nanoclusters (ANCs) within the ES surface have been developed that exhibit superior co-delivery characteristics and produce catalase that enhances HS-PDT efficacy. The unique structure of A/A-ES enables them to co-deliver ALA and ANCs into the HS tissue and to efficiently decompose the endogenous hydrogen peroxide in the HS to generate oxygen. The findings from in vitro and in vivo experiments demonstrated that A/A-ES efficiently co-delivered ALA and ANCs into the HS tissue and that they improved the hypoxic microenvironment of the HS. Systematic assessments reveal that A/A-ES enhance HS-PDT efficacy and that they are highly effective at improving the morphology and promoting HS fibroblast apoptosis and the rearrangement of collagen. These works give rise to an effective treatment option for HSs that integrates the transdermal co-delivery of ALA and nanoenzymes, thereby enabling them to exert their respective beneficial effects, and they highlight the enhancement of HS-PDT efficacy via self-generating oxygen.


Subject(s)
Aminolevulinic Acid/pharmacology , Cicatrix, Hypertrophic/drug therapy , Gold/pharmacology , Nanoparticles/chemistry , Oxygen/metabolism , Photochemotherapy , Photosensitizing Agents/pharmacology , Aminolevulinic Acid/chemistry , Animals , Apoptosis/drug effects , Cells, Cultured , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Drug Delivery Systems , Gold/chemistry , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/metabolism , Particle Size , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Rabbits , Surface Properties
18.
Bioorg Chem ; 109: 104667, 2021 04.
Article in English | MEDLINE | ID: mdl-33611140

ABSTRACT

Photodynamic therapy (PDT) is a promising approach for the targeted treatment of cancer and various other human disorders. An effective, clinically approved approach in PDT involves the administration of 5-aminolevulinic acid (ALA) to generate elevated levels of the natural photosensitiser protoporphyrin IX (PpIX). The development of prodrugs of ALA is of considerable interest as a means to enhance the efficiency and cell selectivity of PpIX accumulation for PDT applications. In this work a novel peptide-targeted dendrimeric prodrug of 5-aminolevulinic acid (ALA) 13 was synthesised which displays nine copies of ALA on a core structure that is linked to a homing peptide for targeted delivery to a specific cancer cell type. The synthesis was accomplished effectively via a flexible, modular solid phase and solution phase route, using a combination of solid phase peptide synthesis and copper-catalysed azide-alkyne cycloaddition chemistry. The prodrug system shows a sustained and enhanced production of protoporphyrin IX (PpIX) in the MDA-MB-231 cell line that over-expresses the epidernal growth factor receptor (EGFR+) in comparison to equimolar ALA and the corresponding non-targeted ALA dendrimer (nine copies of ALA). This study provides a proof of concept for the development of a new generation of prodrugs for ALA-based photodynamic therapy that can deliver an enhanced ALA payload to specific tissue types.


Subject(s)
Aminolevulinic Acid/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Prodrugs , Protoporphyrins/metabolism , Aminolevulinic Acid/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Drug Design , Humans , Molecular Structure , Photosensitizing Agents/chemistry , Structure-Activity Relationship
19.
J Photochem Photobiol B ; 213: 112048, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33142214

ABSTRACT

Iron availability to cells may be modified in the tumour microenvironment, which may be involved in treatment response. Iron availability affects the conversion of protoporphyrin IX to heme, which likely determines the efficacy of aminolevulinic acid-based photodynamic therapy (ALA-based PDT). We compared photoinactivation efficacy in three oesophageal cell lines in culture media differing in iron content, DMEM and RPMI 1640, and in RPMI 1640 supplemented with iron to understand the importance of iron presence for ALA-based PDT outcome. ALA-based PDT was more efficacious in DMEM than in RPMI 1640 in all tested cell lines. Consistently, the highest protoporphyrin IX fluorescence signals, indicating the highest level of protoporphyrin IX production, were detected from cell colonies incubated in DMEM compared to those incubated in RPMI 1640 irrespective of iron presence. Components in the culture media other than iron ions are likely to be responsible for the observed differences in two culture media. Nevertheless, iron supplementation to RPMI 1640 showed that the presence of ferric ions in the concentration range 0-8 mg/l affected ALA-based PDT efficacy in a cell type-dependent manner. In poorly differentiated carcinoma cells, the increased efficacy of ALA-induced photoinactivation in the presence of 0.1 mg/l of supplemented iron was found. At the same iron concentration, the slightly different mitochondrial potential at no modifications of the iron labile pool was observed. The efficacy of ALA-based PDT in vitro depends on the choice of culture medium and the presence of iron ions in culture medium depending on intrinsic properties of cells.


Subject(s)
Aminolevulinic Acid/chemistry , Culture Media/chemistry , Iron/chemistry , Photosensitizing Agents/chemistry , Aminolevulinic Acid/metabolism , Cell Line , Heme/chemistry , Humans , Iron/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Photochemotherapy , Photosensitizing Agents/metabolism , Protoporphyrins/chemistry , Spectrometry, Fluorescence
20.
J Photochem Photobiol B ; 211: 112017, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32919173

ABSTRACT

Aminolevulinic acid (ALA) has been approved as an intraoperative molecular imaging probe for protoporphyrin IX (PpIX) fluorescence-guided resection of glioma. Here we explored its potential application for renal cell carcinoma (RCC) that is showing increased incidence in recent years. ALA-mediated PpIX in cell lysates (intracellular) and culture medium was measured in five human RCC cell lines (786-O, 769-P, A-704, Caki-1, Caki-2) and a non-tumor human kidney epithelial cell line HK-2 by spectrofluorometry and flow cytometry. The activity of PpIX bioconversion enzyme ferrochelatase (FECH) and PpIX efflux transporter ABCG2 was determined to correlate with the PpIX level. We found that ALA-PpIX fluorescence was highly variable among RCC cell lines and A-704 was the only RCC cell line exhibiting significantly higher intracellular PpIX than HK-2 cells. Neither the intracellular PpIX level nor the total amount of PpIX (including PpIX in cell lysates and the medium) had significant correlation with the activity of FECH or ABCG2. To enhance the intracellular PpIX, cells were treated with Ko143, a pharmacological inhibitor of ABCG2. Ko143 significantly increased the intracellular PpIX in cell lines with ABCG2 activity, but not in cell lines with little ABCG2 activity. In fact, there was a positive correlation between the ABCG2 activity and Ko143-induced PpIX enhancement across kidney cell lines. To identify clinically relevant ABCG2 inhibitors, small molecule inhibitors targeting various cell signaling pathways, some of which are known to inhibit ABCG2, were evaluated for the enhancement of ALA-PpIX in Caki-2 cells that had the highest ABCG2 activity in the RCC cell panel. Our screening led to the identification of several clinically available inhibitors that significantly increased the intracellular PpIX. Particularly, kinase inhibitor lapatinib exhibited the strongest enhancement effect. These clinical inhibitors can be used for the enhancement of ALA-PpIX fluorescence in tumors with elevated ABCG2 activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Aminolevulinic Acid/chemistry , Antineoplastic Agents/chemistry , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/therapy , Photosensitizing Agents/chemistry , Protoporphyrins/chemistry , Antineoplastic Agents/pharmacology , Biological Transport , Cell Line, Tumor , Drug Screening Assays, Antitumor , Ferrochelatase/metabolism , Humans , Lapatinib/chemistry , Photochemotherapy , Photosensitizing Agents/pharmacology , Protoporphyrins/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...