Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
Curr Opin HIV AIDS ; 19(2): 56-61, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38169429

ABSTRACT

PURPOSE OF REVIEW: HIV requires lifelong antiviral treatment due to the persistence of a reservoir of latently infected cells. Multiple strategies have been pursued to promote the death of infected cells. RECENT FINDINGS: Several groups have focused on multipronged approaches to induce apoptosis of infected cells. One approach is to combine latency reversal agents with proapoptotic compounds and cytotoxic T cells to first reactivate and then clear infected cells. Other strategies include using natural killer cells or chimeric antigen receptor cells to decrease the size of the reservoir.A novel strategy is to promote cell death by pyroptosis. This mechanism relies on the activation of the caspase recruitment domain-containing protein 8 (CARD8) inflammasome by the HIV protease and can be potentiated by nonnucleoside reverse transcriptase inhibitors. SUMMARY: The achievement of a clinically significant reduction in the size of the reservoir will likely require a combination strategy since none of the approaches pursued so far has been successful on its own in clinical trials. This discrepancy between promising in vitro findings and modest in vivo results highlights the hurdles of identifying a universally effective strategy given the wide heterogeneity of the HIV reservoirs in terms of tissue location, capability to undergo latency reversal and susceptibility to cell death.


Subject(s)
HIV Infections , HIV-1 , Humans , Virus Latency , CD4-Positive T-Lymphocytes , HIV-1/physiology , Cell Death , Neoplasm Proteins/metabolism , Neoplasm Proteins/pharmacology , Neoplasm Proteins/therapeutic use , CARD Signaling Adaptor Proteins/metabolism
2.
Fundam Clin Pharmacol ; 38(2): 351-368, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37793994

ABSTRACT

BACKGROUND: Valproate inhibits clearance of lamotrigine and greatly increases its concentrations. We assessed whether this effect was moderated by a polymorphism (ABCG2 c.421C>A) of the breast cancer resistance protein. METHODS: In two consecutive independent studies in adults with epilepsy on lamotrigine monotherapy or cotreated with valproate: (i) Exposure to valproate was considered treatment, (ii) dose-adjusted lamotrigine troughs at steady state were the outcome, and (iii) ABCG2 c.421C>A genotype (wild-type [wt] homozygosity or variant carriage) was the tested moderator. We used entropy balancing (primary analysis) and exact/optimal full matching (secondary analysis) to control for confounding, including polymorphisms (and linked polymorphisms) suggested to affect exposure to lamotrigine (UGT1A4*3 c.142T>G, rs2011425; UGT2B7-161C>T, rs7668258; ABCB1 1236C>T, rs1128503) to generate frequentist and Bayesian estimates of valproate effects (geometric means ratios [GMR]). RESULTS: The two studies yielded consistent results (replicated); hence, we analyzed combined data (total N = 471, 140 treated, 331 controls, 378 ABCG2 c.421C>A wt subjects, 93 variant carriers). Primary analysis: in variant carriers, valproate effect (GMR) on lamotrigine (treated, n = 21 vs. controls, n = 72) was around 60% higher than in wt subjects (treated, n = 119 vs. controls, n = 259)-ratio of GMRs 1.61 (95%CI 1.23-2.11) (frequentist) and 1.63 (95%CrI 1.26-2.10) (Bayes). Similar differences in valproate effects between variant carriers and wt subjects were found in the secondary analysis (valproate troughs up to 364 µmol/L vs. no valproate; or valproate ≥364 µmol/L vs. no valproate). Susceptibility of the estimates to unmeasured confounding was low. CONCLUSION: Data suggest that polymorphism rs2231142 moderates the effect of valproate on exposure to lamotrigine.


Subject(s)
Breast Neoplasms , Epilepsy , Adult , Humans , Female , Valproic Acid/therapeutic use , Valproic Acid/pharmacology , Lamotrigine/therapeutic use , Bayes Theorem , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/therapeutic use , Epilepsy/drug therapy , Epilepsy/genetics , Anticonvulsants/adverse effects , Breast Neoplasms/drug therapy , Polymorphism, Single Nucleotide
3.
Drug Res (Stuttg) ; 73(8): 473-475, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37591260

ABSTRACT

Bardoxolone methyl (BX) is expected to be an innovate therapeutic agent for chronic kidney disease (CKD). The aim of the present study was to examine whether the expression of subtypes of cytochrome P450 (CYP) and ABC transporters was altered in human hepatocellular carcinoma HepG2 cells by exposure to BX. The expression of mRNAs for CYP1A2, CYP2E1, P-glycoprotein, multidrug resistance-associated protein 1-3, and breast cancer resistance protein was significantly increased by exposure of HepG2 cells to BX, while the expression of CYP3A4 mRNA was significantly decreased under the same conditions. BX had no significant effect on the expression of mRNAs for CYP2C9 and CYP2C19 in HepG2 cells. In conclusion, this study demonstrated that the gene expression of several CYPs and ABC transporters in HepG2 cells was altered when exposed to BX, suggesting the need to pay careful attention to drug-drug interactions in patients receiving BX for CKD treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Renal Insufficiency, Chronic , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Hep G2 Cells , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/therapeutic use , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Gene Expression
4.
Adv Immunol ; 157: 59-100, 2023.
Article in English | MEDLINE | ID: mdl-37061288

ABSTRACT

The biggest challenge to immune control of HIV infection is the rapid within-host viral evolution, which allows selection of viral variants that escape from T cell and antibody recognition. Thus, it is impossible to clear HIV infection without targeting "immutable" components of the virus. Unlike the adaptive immune system that recognizes cognate epitopes, the CARD8 inflammasome senses the essential enzymatic activity of the HIV-1 protease, which is immutable for the virus. Hence, all subtypes of HIV clinical isolates can be recognized by CARD8. In HIV-infected cells, the viral protease is expressed as a subunit of the viral Gag-Pol polyprotein and remains functionally inactive prior to viral budding. A class of anti-HIV drugs, the non-nucleoside reverse transcriptase inhibitors (NNRTIs), can promote Gag-pol dimerization and subsequent premature intracellular activation of the viral protease. NNRTI treatment triggers CARD8 inflammasome activation, which leads to pyroptosis of HIV-infected CD4+ T cells and macrophages. Targeting the CARD8 inflammasome can be a potent and broadly effective strategy for HIV eradication.


Subject(s)
Anti-HIV Agents , HIV Infections , HIV-1 , Humans , Inflammasomes , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcriptase Inhibitors/therapeutic use , Neoplasm Proteins/pharmacology , Neoplasm Proteins/therapeutic use , CARD Signaling Adaptor Proteins
5.
Drug Discov Today ; 28(5): 103537, 2023 05.
Article in English | MEDLINE | ID: mdl-36801375

ABSTRACT

Of the many multidrug resistance (MDR) mechanisms, the ATP binding cassette (ABC) transporters expelling drug molecules out of cells is a major culprit in limiting the efficacy of present-day anticancer drugs. The present review offers an updated information on structure, function, and regulatory mechanisms of major MDR related ABC transporters such as P-glycoprotein, MRP1, BCRP and effect of modulators on their functions. An attempt has been made to provide a focused information on different modulators of ABC transporters so as utilize them in clinical practice for amelioration of emerging MDR crisis in cancer treatment. Finally, the importance ABC transporters as therapeutic targets has been discussed in the light of future strategic planning for translating the ABC transporter inhibitors in clinical practice.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , ATP-Binding Cassette Transporters/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/pharmacology , Multidrug Resistance-Associated Proteins/therapeutic use , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Drug Resistance, Neoplasm , Neoplasm Proteins/metabolism , Neoplasm Proteins/pharmacology , Neoplasm Proteins/therapeutic use , Drug Resistance, Multiple , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Adenosine Triphosphate
6.
J Fluoresc ; 33(3): 1227-1236, 2023 May.
Article in English | MEDLINE | ID: mdl-36811696

ABSTRACT

Recently, nanocarriers have been made to eliminate the disadvantages of chemotherapeutic agents by nanocarriers. Nanocarriers show their efficacy through their targeted and controlled release. In this study, 5-fluorouracil (5FU) was loaded into ruthenium (Ru)-based nanocarrier (5FU-RuNPs) for the first time to eliminate the disadvantages of 5FU, and its cytotoxic and apoptotic effects on HCT116 colorectal cancer cells were compared with free 5FU. 5FU-RuNPs with a size of approximately 100 nm showed a 2.61-fold higher cytotoxic effect compared to free 5FU. Apoptotic cells were detected by Hoechst/propidium iodide double staining, and the expression levels of BAX/Bcl-2 and p53 proteins, in which apoptosis occurred intrinsically, were revealed. In addition, 5FU-RuNPs was also found to reduce multidrug resistance (MDR) according to BCRP/ABCG2 gene expression levels. When all the results were evaluated, the fact that Ru-based nanocarriers alone did not cause cytotoxicity proved that they were ideal nanocarriers. Moreover, 5FU-RuNPs did not show any significant effect on the cell viability of normal human epithelial cell lines BEAS-2B. Consequently, the 5FU-RuNPs synthesized for the first time may be ideal candidates for cancer treatment because they can minimize the potential drawbacks of free 5FU.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Ruthenium , Humans , Ruthenium/pharmacology , Ruthenium/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Neoplasm Proteins/metabolism , Neoplasm Proteins/pharmacology , Neoplasm Proteins/therapeutic use , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , HCT116 Cells , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cell Line, Tumor
7.
Drug Res (Stuttg) ; 73(3): 146-155, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36630991

ABSTRACT

Imatinib mesylate (IM) is the gold standard for treatment of Chronic Myeloid Leukemia (CML). This study aimed to gain more knowledge of the altered PK, pharmacogenetic factors, and gene expression leading to variable IM levels. Fifty patients with chronic phase-CML were enrolled in this study and divided as 25 responders and 25 non-responders (patients are directly recruited after response assessment). HPLC/MS/MS was used to determine trough and peak concentration of imatinib and N-desmethyl imatinib in the blood. PCR-RFLP technique was used to detect IDH1 gene mutation (R132). The median value of IM trough level was significantly higher, the P/T ratio was significantly lower and the α-1-acid glycoprotein (AGP) was significantly higher among responders compared to non-responders (P=0.007, 0.009 and 0.048, respectively). Higher N-desmethyl imatinib peak plasma concentration was observed with low mRNA expression of ABCG2 and OCT1 (P=0.01 and 0.037, respectively). IDH1 R132 gene mutation was associated with a significant increase in toxicities (P=0.028). In conclusion, IM trough level, P/T ratio and AGP was significantly higher in responders. In addition, ABCG2 and OCT1 gene expression may affect the interindividual PK variation. Although a prospective study with a larger patient population is necessary to validate these findings. IDH1 mutation is a predictor of increased toxicity with IM treatment.


Subject(s)
Antineoplastic Agents , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Humans , Imatinib Mesylate/adverse effects , Tandem Mass Spectrometry , Egypt , Prospective Studies , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mutation , Antineoplastic Agents/adverse effects , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/therapeutic use , Isocitrate Dehydrogenase/genetics
8.
Expert Rev Clin Pharmacol ; 16(2): 133-148, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36708283

ABSTRACT

INTRODUCTION: FLT3 inhibitors (FLT3i) are drugs in which there is limited experience and not yet enough information on the mechanisms of absorption, transport, and elimination; but especially on the potential drug-drug interactions (DDIs). There are therefore risks in the management of FLT3i DDIs (i.e. sorafenib, ponatinib, crenolanib, midostaurin, quizartinib, and gilteritinib) and ignoring them can compromise therapeutic success in acute myeloid leukemia (AML) treatment, in complex patients and secondary pathologies. AREAS COVERED: This review summarizes the DDIs of FLT3i with P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), organic anion transporting (OAT), organic cationic transporting (OCT), cytochrome P450 (CYP) subunits, and other minor metabolic/transport pathways. EMBASE, PubMed, the Cochrane Central Register and the Web of Science were searched. The last literature search was performed on the 14 February 2022. EXPERT OPINION: FLT3i will be combined with other therapeutic agents (supportive care, doublet, or triplet therapy) and in different clinical settings, which means a greater chance of controlling and even eradicating the disease effectively, but also an increased risk to patients due to potential DDIs. Healthcare professionals should be aware of the potential interactions that may occur and be vigilant in monitoring those patients who are receiving any potentially interacting drug.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Humans , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Antineoplastic Agents/adverse effects , Neoplasm Proteins/therapeutic use , Protein Kinase Inhibitors/adverse effects , Drug Interactions , Leukemia, Myeloid, Acute/drug therapy , fms-Like Tyrosine Kinase 3 , Mutation
9.
Chemosphere ; 312(Pt 1): 136545, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36155021

ABSTRACT

OBJECTIVE: It has been suggested that Bisphenol A (BPA), a high-production-volume industrial chemical, can accelerate the development of various type of cancers. However, the effect of BPA on osteosarcoma and the underlying mechanisms are yet to be established. Therefore, in this study we tried to explore the carcinogenic effects of BPA on osteosarcoma and the underlying mechanism. METHODS: SaOs-2 cancer cell line was used to treat with BPA at the doses of 0.1, 1, 10 µM DGLAP5 knockdown and overexpression methods were constructed by using adenovirus mediated transfection, and the functional analysis of DGLAP5 was investigated to evaluate the carcinogenic effect of BPA on osteosarcoma through DLGAP5. Xenograft and metastatic mouse model were used to evaluate in vivo experiments. RESULTS: In this study, BPA at 10 µM promoted the proliferation, migration and invasion in vitro, and accelerate the progression and metastasis in vivo. Also, exposure to BPA was associated with poor survival of osteosarcoma in mice. In addition, we observed that BPA at 10 µM significantly increased the expression of DGLAP5 in osteosarcoma. Silencing DGLAP5 could reverse the effect of BPA on proliferation, migration and invasion. Mechanically, BPA promoted IL-6, JAK2, and STAT3 expression and promoted tumor progression in an IL-6-dependent manner through up-regulation of DLGAP5. CONCLUSION: Our findings demonstrated that BPA could promote the proliferation, migration, invasion of osteosarcoma cells and related to poor survival in a mouse model. DLGAP5 is one of the most critical targets of BPA to act as a carcinogen through IL-6/JAK2/STAT3 signaling pathway.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Mice , Animals , Interleukin-6/genetics , Interleukin-6/metabolism , Cell Movement , Osteosarcoma/chemically induced , Osteosarcoma/genetics , Signal Transduction , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Bone Neoplasms/chemically induced , Bone Neoplasms/genetics , Cell Proliferation , Cell Line, Tumor , Neoplasm Proteins/metabolism , Neoplasm Proteins/pharmacology , Neoplasm Proteins/therapeutic use , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Janus Kinase 2/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
10.
RMD Open ; 8(1)2022 05.
Article in English | MEDLINE | ID: mdl-35589331

ABSTRACT

OBJECTIVES: Given the similarity in symptoms between primary Sjogren's syndrome (SjS) and non-SjS sicca syndrome (sicca), we sought to characterise clinical and proteomic predictors of symptoms in both groups in order to better understand disease mechanisms and help guide development of immunomodulatory treatments. These have not, to date, unequivocally improved symptoms in SjS clinical trials. METHODS: Serum proteomics was performed using O-link inflammation and cardiovascular II panels. SjS (n=53) fulfilled 2016 ACR/European Alliance of Associations for Rheumatology (EULAR) criteria whereas sicca (n=60) were anti-Ro negative, displayed objective or subjective dryness, and either had a negative salivary gland biopsy or, in the absence of a biopsy, it was considered that a biopsy result would not change classification status. Linear regression analysis was performed to identify the key predictors of symptoms. Cluster analysis was completed using protein expression values. RESULTS: EULAR-Sjögren's-Syndrome-Patient-Reported-Index (ESSPRI), EuroQoL-5 Dimension utility values, and anxiety and depression did not differ between SjS and sicca. Correlations between body mass index (BMI) and ESSPRI were found in sicca and to a lesser extent in SjS. Twenty proteins positively associated with symptoms in sicca but none in SjS. We identified two proteomically defined subgroups in sicca and two in SjS that differed in symptom burden. Within hierarchical clustering of the SjS and sicca pool, the highest symptom burden groups were the least distinct. Levels of adrenomedullin (ADM), soluble CD40 (CD40) and spondin 2 (SPON2) together explained 51% of symptom variability in sicca. ADM was strongly correlated with ESSPRI (spearman's r=0.62; p<0.0001), even in a multivariate model corrected for BMI, age, objective dryness, depression and anxiety scores. CONCLUSIONS: Obesity-related metabolic factors may regulate symptoms in sicca. Further work should explore non-inflammatory drivers of high symptom burden in SjS to improve clinical trial outcomes.


Subject(s)
Rheumatology , Sjogren's Syndrome , Anxiety/etiology , Extracellular Matrix Proteins/therapeutic use , Humans , Neoplasm Proteins/therapeutic use , Proteomics , Sjogren's Syndrome/complications , Sjogren's Syndrome/diagnosis , Sjogren's Syndrome/drug therapy
11.
Cell Mol Neurobiol ; 42(4): 1005-1020, 2022 May.
Article in English | MEDLINE | ID: mdl-33245508

ABSTRACT

The tumor resistance of glioblastoma cells in vivo is thought to be enhanced by their heterogeneity and plasticity, which are extremely difficult to curb in vitro. The external microenvironment shapes the molecular profile of tumor culture models, thus influencing potential therapy response. Our study examines the expression profile of selected lncRNAs involved in tumor resistance network in three different glioblastoma-derived models commonly utilized for testing drug response in vitro. Differential expression analysis revealed significant divergence in lncRNA profile between parental tumors and tumor-derived cell cultures in vitro, including the following particles: MALAT1, CASC2, H19, TUSC7, XIST, RP11-838N2.4, DLX6-AS1, GLIDR, MIR210HG, SOX2-OT. The examined lncRNAs influence the phenomenon of tumor resistance via their downstream target genes through a variety of processes: multi-drug resistance, epithelial-mesenchymal transition, autophagy, cell proliferation and viability, and DNA repair. A comparison of in vivo and in vitro expression identified differences in the levels of potential lncRNA targets, with the highest discrepancies detected for the MDR1, LRP1, BCRP and MRP1 genes. Co-expression analyses confirmed the following interrelations: MALAT1-TYMS, MALAT1-MRP5, H19-ZEB1, CASC2-VIM, CASC2-N-CAD; they additionally suggest the possibility of MALAT1-BCRP, MALAT1-mTOR and TUSC7-PTEN interconnections in glioblastoma. Although our results clearly demonstrate that the artificial ex vivo microenvironment changes the profile of lncRNAs related to tumor resistance, it is difficult to anticipate the final phenotypic effect, since this phenomenon is a complex one that involves a network of molecular interactions underlying a variety of cellular processes.


Subject(s)
Glioblastoma , RNA, Long Noncoding , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Cell Culture Techniques , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/therapeutic use , RNA, Long Noncoding/genetics , Tumor Microenvironment
12.
Epilepsia Open ; 7 Suppl 1: S34-S46, 2022 08.
Article in English | MEDLINE | ID: mdl-34542938

ABSTRACT

The multidrug-resistance (MDR) phenotype is typically observed in patients with refractory epilepsy (RE) whose seizures are not controlled despite receiving several combinations of more than two antiseizure medications (ASMs) directed against different ion channels or neurotransmitter receptors. Since the use of bromide in 1860, more than 20 ASMs have been developed; however, historically ~30% of cases of RE with MDR phenotype remains unchanged. Irrespective of metabolic biotransformation, the biodistribution of ASMs and their metabolites depends on the functional expression of some ATP-binding cassette transporters (ABC-t) in different organs, such as the blood-brain barrier (BBB), bowel, liver, and kidney, among others. ABC-t, such as P-glycoprotein (P-gp), multidrug resistance-associated protein (MRP-1), and breast cancer-resistance protein (BCRP), are mainly expressed in excretory organs and play a critical role in the pharmacokinetics (PK) of all drugs. The transporter hypothesis can explain pharmacoresistance to a broad spectrum of ASMs, even when administered simultaneously. Since ABC-t expression can be induced by hypoxia, inflammation, or seizures, a high frequency of uncontrolled seizures increases the risk of RE. These stimuli can induce ABC-t expression in excretory organs and in previously non-expressing (electrically responsive) cells, such as neurons or cardiomyocytes. In this regard, an alternative mechanism to the classical pumping function of P-gp indicates that P-gp activity can also produce a significant reduction in resting membrane potential (ΔΨ0 = -60 to -10 mV). P-gp expression in neurons and cardiomyocytes can produce membrane depolarization and participate in epileptogenesis, heart failure, and sudden unexpected death in epilepsy. On this basis, ABC-t play a peripheral role in controlling the PK of ASMs and their access to the brain and act at a central level, favoring neuronal depolarization by mechanisms independent of ion channels or neurotransmitters that current ASMs cannot control.


Subject(s)
Epilepsy , Neoplasm Proteins , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/therapeutic use , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/therapeutic use , Epilepsy/drug therapy , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/therapeutic use , Seizures/drug therapy , Tissue Distribution
13.
Biochim Biophys Acta Rev Cancer ; 1876(2): 188635, 2021 12.
Article in English | MEDLINE | ID: mdl-34656686

ABSTRACT

The involvement of the Gasdermin (GSDM) protein family in cancer and other pathologies is one of the hottest topics in biomedical research. There are six GSDMs in humans (GSDMA, B, C, D, GSDME/DFNA5 and PJVK/DFNB59) and, except PJVK, they can trigger cell death mostly by pyroptosis (a form of lytic and pro-inflammatory cell death) but also other mechanisms. The exact role of GSDMs in cancer is intricate, since depending on the biological context, these proteins have diverse cell-death dependent and independent functions, exhibit either pro-tumor or anti-tumor functions, and promote either sensitization or resistance to oncologic treatments. In this review we provide a comprehensive overview on the multifaceted roles of the GSDMs in cancer, and we critically discuss the possibilities of exploiting GSDM functions as determinants of anti-cancer treatment and as novel therapeutic targets, with special emphasis on innovative GSDM-directed nano-therapies. Finally, we discuss the issues to be resolved before GSDM-mediated oncologic therapies became a reality at the clinical level.


Subject(s)
Biology , Medical Oncology/methods , Neoplasm Proteins/therapeutic use , Neoplasms/therapy , Humans , Neoplasm Proteins/pharmacology
14.
Gut Microbes ; 13(1): 1946369, 2021.
Article in English | MEDLINE | ID: mdl-34275417

ABSTRACT

Gasdermin D (GSDMD) functions as a key pyroptotic executor through its secreted N-terminal domain (GSDMD-N). However, the functional relevance and mechanistic basis of the precise roles of host colonic GSDMD in high-fat diet (HFD)-induced gut dysbiosis and systemic endotoxemia remain elusive. In this study, we demonstrate that HFD feeding triggers GSDMD-N secretion of both T-lymphocytes and enterocytes in mouse colons. GSDMD deficiency aggravates HFD-induced systemic endotoxemia, gut barrier impairment, and colonic inflammation. More importantly, active GSDMD-N kills the Proteobacteria phylum via directly interacting with Cardiolipin. Mechanistically, we identify that the Glu236 (a known residue for GSDMD protein cleavage) is a bona fide important site for the bacterial recognition of GSDMD. Collectively, our findings explain the mechanism by which colonic GSDMD-N maintains low levels of HFD-induced metabolic endotoxemia. A GSDMD-N mimetic containing an exposed Glu236 site could be an attractive strategy for the treatment of HFD-induced metabolic endotoxemia.


Subject(s)
Colon/microbiology , Dysbiosis/chemically induced , Dysbiosis/drug therapy , Endotoxemia/chemically induced , Endotoxemia/drug therapy , Neoplasm Proteins/pharmacokinetics , Neoplasm Proteins/therapeutic use , Proteobacteria/drug effects , Animals , Cardiolipins/analysis , Diet, High-Fat/adverse effects , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Humans , Mice
15.
Biomolecules ; 11(6)2021 05 22.
Article in English | MEDLINE | ID: mdl-34067242

ABSTRACT

Oximes have been studied for decades because of their significant roles as acetylcholinesterase reactivators. Over the last twenty years, a large number of oximes have been reported with useful pharmaceutical properties, including compounds with antibacterial, anticancer, anti-arthritis, and anti-stroke activities. Many oximes are kinase inhibitors and have been shown to inhibit over 40 different kinases, including AMP-activated protein kinase (AMPK), phosphatidylinositol 3-kinase (PI3K), cyclin-dependent kinase (CDK), serine/threonine kinases glycogen synthase kinase 3 α/ß (GSK-3α/ß), Aurora A, B-Raf, Chk1, death-associated protein-kinase-related 2 (DRAK2), phosphorylase kinase (PhK), serum and glucocorticoid-regulated kinase (SGK), Janus tyrosine kinase (JAK), and multiple receptor and non-receptor tyrosine kinases. Some oximes are inhibitors of lipoxygenase 5, human neutrophil elastase, and proteinase 3. The oxime group contains two H-bond acceptors (nitrogen and oxygen atoms) and one H-bond donor (OH group), versus only one H-bond acceptor present in carbonyl groups. This feature, together with the high polarity of oxime groups, may lead to a significantly different mode of interaction with receptor binding sites compared to corresponding carbonyl compounds, despite small changes in the total size and shape of the compound. In addition, oximes can generate nitric oxide. This review is focused on oximes as kinase inhibitors with anticancer and anti-inflammatory activities. Oximes with non-kinase targets or mechanisms of anti-inflammatory activity are also discussed.


Subject(s)
Anti-Inflammatory Agents , Antineoplastic Agents , Enzyme Inhibitors , Neoplasms/drug therapy , Oximes , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/therapeutic use , Neoplasms/enzymology , Oximes/chemistry , Oximes/therapeutic use , Phosphotransferases/antagonists & inhibitors , Phosphotransferases/metabolism
16.
Clin Transl Oncol ; 23(8): 1688-1704, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33792840

ABSTRACT

Osteosarcoma, a common malignant tumor in orthopedics, often has a very poor prognosis after lung metastasis. Immunotherapy has not achieved much progress in the treatment because of the characteristics of solid tumors and immune environment of osteosarcoma. The tumor environment is rather essential for sarcoma treatment. Our previous study demonstrated that heat shock proteins could be used as antitumor vaccines by carrying tumor antigen peptides, and we hypothesize that an anti-osteosarcoma effect may be increased with an immune check point inhibitor (PD-L1 inhibitor) as a combination treatment strategy. The present study prepared a multisubtype mixed heat shock protein osteosarcoma vaccine (mHSP/peptide vaccine) and concluded that the mHSP/peptide vaccine was more effective than a single subtype heat shock protein, like Grp94. Therefore, we used the mHSP/peptide vaccine in combination with a PD-L1 inhibitor to treat osteosarcoma, and the deterioration of osteosarcoma was effectively hampered. The mechanism of combined therapy was investigated, and AKT expression participates with sarcoma lung metastasis. This study proposed an antisarcoma strategy via stimulation of the immune system as a further alternative approach for sarcoma treatment and elucidated the mechanism of combined therapy.


Subject(s)
Bone Neoplasms/therapy , Cancer Vaccines/therapeutic use , Heat-Shock Proteins/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Osteosarcoma/therapy , Animals , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Cell Line, Tumor , Combined Modality Therapy/methods , Disease Models, Animal , Female , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunotherapy/methods , Interferon-gamma/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Neoplasm Proteins/immunology , Neoplasm Proteins/therapeutic use , Osteosarcoma/genetics , Osteosarcoma/immunology , Osteosarcoma/secondary , Proto-Oncogene Proteins c-akt/metabolism , Tumor Escape
17.
Adv Skin Wound Care ; 34(4): 1-7, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33739952

ABSTRACT

OBJECTIVE: To discuss human amnion chorion (placental) membrane allograft (HACMA) use for the treatment of chronic diabetic foot ulcers (DFUs) and to evaluate the effectiveness, cost, and product waste of this therapy. DATA SOURCES: PubMed, Cochrane, and OVID databases. STUDY SELECTION: Twenty-four articles pertaining to HACMA and DFUs published from 2016 to 2020 were selected. DATA EXTRACTION: The data collected included type of wound care product, study design, study size, baseline size of DFU, cost, product wastage, number of applications, and wound healing outcomes. DATA SYNTHESIS: Human amnion chorion membrane allografts in the treatment of chronic DFUs have led to a reduction in healing time and increased the overall percentage of healing, making them more effective in treating DFUs compared with standard of care. These products are offered in multiple sizes with various shelf lives and methods of storage, making them accessible, easy to use, less wasteful, and lower in cost compared with other commercially available products. Promising evidence demonstrates that HACMAs are beneficial in treating complex, high-grade DFUs with exposed tendon or bone. CONCLUSIONS: Human amnion chorion membrane allografts are effective in treating chronic DFUs with a greater percentage of complete wound closure and a reduction in healing time versus standard of care.


Subject(s)
Allografts/standards , Cysteine Endopeptidases/pharmacology , Diabetic Foot/surgery , Neoplasm Proteins/pharmacology , Allografts/statistics & numerical data , Amnion/transplantation , Chorion/transplantation , Cysteine Endopeptidases/therapeutic use , Humans , Neoplasm Proteins/therapeutic use , Treatment Outcome
18.
Cells ; 9(11)2020 11 23.
Article in English | MEDLINE | ID: mdl-33238381

ABSTRACT

Although papillary thyroid carcinoma (PTC) has a good prognosis, 20-90% of patients show metastasis to regional lymph nodes and 10-15% of patients show metastasis to distant sites. Metastatic disease represents the main clinical challenge that impacts survival rate. We previously showed that LIMD2 was a novel metastasis-associated gene. In this study, to interrogate the role of LIMD2 in cancer invasion and metastasis, we used CRISPR-mediated knockout (KO) of LIMD2 in PTC cells (BCPAP and TPC1). Western blot and high-content screening (HCS) analysis confirmed functional KO of LIMD2. LIMD2 KO reduced in vitro invasion and migration. Ultrastructural analyses showed that cell polarity and mitochondria function and morphology were restored in LIMD2 KO cells. To unveil the signals supervising these phenotypic changes, we employed phospho-protein array. Several members of the MAPK superfamily showed robust reduction in phosphorylation. A Venn diagram displayed the overlap of kinases with reduced phosphorylation in both cell lines and showed that they were able to initiate or sustain the epithelial-mesenchymal transition (EMT) and DNA damage checkpoint. Flow cytometry and HCS validation analyses further corroborated the phospho-protein array data. Collectively, our findings show that LIMD2 enhances phosphorylation of kinases associated with EMT and invasion. Through cooperation with different kinases, it contributes to the increased genomic instability that ultimately promotes PTC progression.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Proteins/therapeutic use , Thyroid Cancer, Papillary/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Male , Neoplasm Metastasis , Neoplasm Proteins/pharmacology , Thyroid Cancer, Papillary/pathology
19.
Biosci Rep ; 40(12)2020 12 23.
Article in English | MEDLINE | ID: mdl-33169789

ABSTRACT

Immunotherapy is a breakthrough approach for cancer treatment and prevention. By exploiting the fact that cancer cells have overexpression of tumor antigens responsible for its growth and progression, which can be identified and removed by boosting the immune system. In silico techniques have provided efficient ways for developing preventive measures to ward off cancer. Herein, we have designed a potent cytotoxic T-lymphocyte epitope to elicit a desirable immune response against carcinogenic melanoma-associated antigen-A11. Potent epitope was predicted using reliable algorithms and characterized by advanced computational avenue CABS molecular dynamics simulation, for full flexible binding with HLA-A*0201 and androgen receptor to large-scale rearrangements of the complex system. Results showed the potent immunogenic construct (KIIDLVHLL), from top epitopes using five algorithms. Molecular docking analyses showed the strong binding of epitope with HLA-A*0201 and androgen receptor with docking score of -780.6 and -641.06 kcal/mol, respectively. Molecular dynamics simulation analysis revealed strong binding of lead epitope with androgen receptor by involvement of 127 elements through atomic-model study. Full flexibility study showed stable binding of epitope with an average root mean square deviation (RMSD) 2.21 Å and maximum RMSD value of 6.48 Å in optimal cluster density area. The epitope also showed remarkable results with radius of gyration 23.0777 Å, world population coverage of 39.08% by immune epitope database, and transporter associated with antigen processing (TAP) affinity IC50 value of 2039.65 nm. Moreover, in silico cloning approach confirmed the expression and translation capacity of the construct within a suitable expression vector. The present study paves way for a potential immunogenic construct for prevention of cancer.


Subject(s)
Antigens, Neoplasm/therapeutic use , Cancer Vaccines/therapeutic use , Cytotoxicity, Immunologic , Drug Design , Epitopes, T-Lymphocyte , Neoplasm Proteins/therapeutic use , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Algorithms , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Immunogenicity, Vaccine , Molecular Docking Simulation , Molecular Dynamics Simulation , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Receptors, Androgen/immunology , Receptors, Androgen/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/metabolism , Vaccines, Subunit/therapeutic use
20.
Photodiagnosis Photodyn Ther ; 32: 102039, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33017656

ABSTRACT

OBJECTIVE: To investigate the effect of 5-aminolevulinic acid (ALA) mediated photodynamic therapy (PDT) on the invasion and metastasis in cutaneous squamous cell carcinoma (cSCC) cell line(SCL-1) and to study whether the effect was via the MTSS1 gene and p63 gene related pathways. METHODS: SCL-1 cells were cultured and submitted to ALA-PDT treatment (ALA-PDT group), ALA treatment alone (ALA group), LED illumination alone (LED group) and remains untreated (control group). Scratch test, Transwell migration chamber assay and Matrigel cell invasion assay were used to detect the ability of migration and invasion of SCL-1 cells after treatment. The mRNA levels and protein expressions of tumor metastasis suppressor gene (MTSS1) and p63 gene were further detected by using quantitative real-time PCR and flow cytometry assay respectively after treatment. RESULTS: The migration and invasion abilities of SCL-1 cells after treatment were significantly reduced in the ALA-PDT groups than that in ALA group, LED group and control group (P<0.05). Both the mRNA and protein expression levels of MTSS1 gene were up-regulated, while the mRNA and protein expression levels of p63 gene were down-regulated after ALA-PDT treatment. CONCLUSION: ALA-PDT suppressed the migration and invasion of human cSCC cell line, probably via the MTSS1 gene and p63 gene related pathways. This study put forward a possible mechanism of invasion in SCL-1 cell, also providing a potential target for the therapy of cSCC.


Subject(s)
Aminolevulinic Acid , Carcinoma, Squamous Cell , Photochemotherapy , Skin Neoplasms , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Humans , Membrane Proteins , Microfilament Proteins/therapeutic use , Neoplasm Proteins/therapeutic use , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...