1
|
Cheng J, Qin X, Han B, Gu H, Zou H, Peng P, Mao Z, Li B. Mechanism of starvation induced autophagy and apoptosis in the midgut of silkworm, Bombyx mori, based on calcium homeostasis. INSECT MOLECULAR BIOLOGY 2025; 34:440-451. [PMID: 39707627 DOI: 10.1111/imb.12981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024]
Abstract
Starvation can induce autophagy and apoptosis in intestinal cells. To elucidate the underlying mechanisms, we investigated autophagy and apoptosis in the midgut of the model insect, silkworm (Bombyx mori), focusing on calcium homeostasis. The results indicated that the body weight of silkworms decreased, along with damage to the morphology of their digestive tracts and midguts after starvation treatment. Additionally, mitochondrial swelling, autophagy and apoptosis were observable. Further investigation revealed that starvation upregulated the transcription of Ca2+ release channel-associated genes (e.g., BmIP3R, BmRyR) but suppressed the expression of Ca2+ efflux genes (BmPMCA), resulting in Ca2+ overload in midgut cells and subsequent upregulation of BmCalpain transcription. In addition, starvation increased the transcription of key autophagy genes (BmATG5, BmATG7, BmATG8) and the expression of the LC3-II protein. Upon prolonged starvation, the NtATG5 protein levels increased, a process that facilitated the transition from autophagy to apoptosis. These results indicate that Ca2+ overload activates the calpain-mediated apoptosis pathway and promotes apoptosis of midgut cells. The present study reveals the significant role that Ca2+ plays in the occurrence and transformation of autophagy and apoptosis induced by starvation treatment, thus providing a new research strategy for investigating the damage caused by starvation in biological organisms.
Collapse
Affiliation(s)
- Jialu Cheng
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Xueling Qin
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Bing Han
- Sericulture Research Institute of Liaoning Province, Dandong, China
| | - Haoyi Gu
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Hongbing Zou
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Peiling Peng
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhongxu Mao
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Bing Li
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, China
- Sericulture Institute of Soochow University, Suzhou, China
| |
Collapse
|
2
|
Zemitis A, Vanags J, Klavins K, Laganovska G. The Role of IDO Activity in Cataract Progression: Correlation to Age and Cataract Severity. Curr Eye Res 2025:1-7. [PMID: 40423999 DOI: 10.1080/02713683.2025.2506118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/28/2025]
Abstract
PURPOSE Cataract is a prevalent, progressive lens disorder characterized by gradual opacity, often related to aging. Oxidative stress, ultraviolet radiation, and inflammation contribute to lens damage and protein modifications. Indoleamine 2,3-dioxygenase plays a critical role in tryptophan metabolism, with kynurenines implicated in various diseases. This study aimed to assess IDO activity in cataract patients by analyzing IDO activity in aqueous humor. METHODS The study included 170 cataract surgery patients (60 males, 110 females; mean age 73.9 ± 9.43 years). Cataract severity was categorized using the SPONCS system: SPONCS 1 (1.8%), SPONCS 2 (30.6%), SPONCS 3 (35.3%), SPONCS 4 (11.8%), and SPONCS 5 (20.6%). Aqueous humor samples (50-120 µL) were collected via paracentesis and analyzed using HILIC liquid chromatography with mass spectrometry. Target metabolite quantification was performed using internal standards and calibration curves. RESULTS Significant differences in IDO activity were observed across SPONCS groups (X2(4) = 12.0, p = .018, Ɛ2= 0.0707), particularly between SPONCS 2 and SPONCS 5 (p = .013). Age differences across SPONCS groups were also significant (p < .001). Males had lower tryptophan levels than females (p = .027). Correlations were found between SPONCS and IDO activity (rs = -0.255, p < .001), SPONCS and kynurenine (rs = 0.196, p = .011), and kynurenic acid with kynurenine (rs = 0.355, p < .001). CONCLUSION Patients with SPONCS 2 cataract exhibit increased susceptibility to elevated IDO activity and heightened kynurenine production. IDO serves as a more reliable prognostic marker for cataract progression than chronological age. Furthermore, IDO activity may be associated with reduced glutathione levels in human lens epithelial cells, suggesting a potential link between the enzyme and oxidative stress within the lens.
Collapse
Affiliation(s)
- Arturs Zemitis
- Department of Ophthalmology, Riga Stradins University, Riga, Latvia
- Clinic of Ophthalmology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Juris Vanags
- Department of Ophthalmology, Riga Stradins University, Riga, Latvia
- Clinic of Ophthalmology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Kristaps Klavins
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Riga, Latvia
| | - Guna Laganovska
- Department of Ophthalmology, Riga Stradins University, Riga, Latvia
- Clinic of Ophthalmology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| |
Collapse
|
3
|
Khairani AF, Shalannandia WA, Bashari MH, Atik N. Aaptamine Alters Vimentin Expression and Migration Capability of Triple-Negative Breast Cancer Cells. J Exp Pharmacol 2025; 17:239-247. [PMID: 40420842 PMCID: PMC12105642 DOI: 10.2147/jep.s512099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/17/2025] [Indexed: 05/28/2025] Open
Abstract
Purpose This study aimed to explore the effect of Aaptamine, an alkaloid derived from marine sponges, on the vimentin expression in both mRNA and protein levels and the migration capacity of breast cancer cells. Methods The triple-negative breast cancer cell line MDA-MB-231 was used for in vitro experiments. Low-cytotoxicity concentrations of Aaptamine (12.5 to 50 μM) were given to MDA-MB-231 cells. The vimentin mRNA and protein expression were evaluated using RT-qPCR and immunofluorescence, respectively, 72 h after Aaptamine treatment. The migration scratch assay was conducted for 48 hours. Results Aaptamine treatment in three different doses did not affect the expression of vimentin at the mRNA level while significantly lowering vimentin protein expression at the concentration of 12.5 µM. In addition, Aaptamine significantly inhibited breast cancer cell migration in a dose-dependent manner. Conclusion Aaptamine inhibits vimentin protein expression and demonstrates anti-migration activity in the sub-cytotoxic dose.
Collapse
Affiliation(s)
- Astrid Feinisa Khairani
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
- Graduate School of Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Widad Aghnia Shalannandia
- Graduate School of Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Muhammad Hasan Bashari
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
- Graduate School of Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Nur Atik
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
- Graduate School of Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| |
Collapse
|
4
|
Wang J, Chu Y, Hu G, Liang S, Cui Y, Zuo J, Luo Z, Chen X, Wang X, Yu Z, Zhang Z, Wang X. Piperlongumine Inhibits Malignant Progression of Esophageal Squamous Cells Through the PI3K/AKT Signaling Pathway. Biochem Genet 2025:10.1007/s10528-025-11139-7. [PMID: 40383869 DOI: 10.1007/s10528-025-11139-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
To examine the impact of Piperlongumine (PL) on the proliferation, migration, invasion, cell cycle progression, and apoptosis in esophageal squamous cell carcinoma (ESCC) cells, as well as to elucidate the underlying molecular mechanisms. The suppressive effects of PL on the viability of ESCC cells were assessed using the CCK-8 assay, bright field imaging, and colony formation assays. Apoptosis induction and cell cycle disruption by PL were evaluated using flow cytometry. The impact of PL on ESCC cell migration and invasion was examined through scratch healing and Transwell assays. Differential gene expression analysis of ESCC tumor and normal tissues from the GSE29886 dataset, integrated with network pharmacology predictions, was conducted to identify core genes and molecular mechanisms involved in PL action. Key protein expression levels in the apoptosis, epithelial-mesenchymal transition (EMT), and PI3K/AKT signaling pathways were quantified by Western blotting. The CCK-8 and colony formation assays demonstrated that PL effectively suppressed cell viability and proliferation in ESCC. Flow cytometry revealed that PL down-regulated CDK1 expression, resulting in G2/M phase arrest, and promoted apoptosis by decreasing Bcl-2 levels and increasing cleaved caspase-3 and PARP. The scratch and Transwell assays indicated that PL inhibited ESCC cell migration and invasion, down-regulated the EMT-associated proteins Vimentin and N-cadherin, and up-regulated E-cadherin. Western blotting confirmed the down-regulation of P-PI3K and P-AKT, indicating the inhibition of the PI3K/AKT pathway by PL. These findings offer a pharmacological foundation for the development of PL as a potential phytotherapeutic agent for the clinical management of ESCC.
Collapse
Affiliation(s)
- Jun Wang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Yueming Chu
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Guangbing Hu
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Shiqi Liang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Yutong Cui
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Ji Zuo
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Zichen Luo
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xinrui Chen
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xiaobo Wang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Zhenghang Yu
- Department of General Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Zhongyang Zhang
- Department of General Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xianfei Wang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
- Digestive Endoscopy Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
5
|
Quintero-Espinosa DA, Velez-Pardo C, Jimenez-Del-Rio M. PF-06447475 Molecule Attenuates the Neuropathology of Familial Alzheimer's and Coexistent Parkinson's Disease Markers in PSEN1 I416T Dopaminergic-like Neurons. Molecules 2025; 30:2034. [PMID: 40363838 PMCID: PMC12074268 DOI: 10.3390/molecules30092034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Familial Alzheimer's disease (FAD) is a complex multifactorial disorder clinically characterized by cognitive impairment and memory loss. Pathologically, FAD is characterized by intracellular accumulation of the protein fragment Aβ42 (iAβ), hyperphosphorylated microtubule-associated protein TAU (p-TAU), and extensive degeneration of basal forebrain cholinergic neurons of the nucleus basalis of Meynert (NbM) and the medial septal nucleus (MSN), mainly caused by mutations in the amyloid precursor protein (APP), presenilin 1 (PSEN1), and PSEN2 gene. Since the dopaminergic system may contribute to FAD symptoms, alterations in the nigro-hippocampal pathway may be associated with cognitive impairment in FAD. Interestingly, p-α-synuclein (p-α-Syn), Aβ, and p-TAU have been found to coexist in vulnerable regions of postmortem AD brains. However, the mechanism by which Aβ, p-TAU, and α-Syn coexist in DAergic neurons in AD brains has not been determined. We generated PSEN1 I416T dopaminergic-like neurons (DALNs) from I416T menstrual stromal cells (MenSCs) in NeuroForsk 2.0 medium for 7 days and then cultured them in minimal culture medium (MCm) for another 4 days. On day 11, DALNs were analyzed for molecular and pathological markers by flow cytometry and fluorescence microscopy. We found that mutant DALNs showed increased accumulation of iAβ as well as increased phosphorylation of TAU at S202/T205 compared to WT DALNs. Thus, mutant DALNs exhibited typical pathological hallmarks of Alzheimer's disease. Furthermore, PSEN1 I416T DALNs showed concomitant signs of OS as evidenced by the appearance of oxidized sensor protein DJ-1 (i.e., DJ-1C106-SO3) and apoptotic markers TP53, pS63-c-JUN, PUMA, and cleavage caspase 3 (CC3). Notably, these DALNs exhibited PD-associated proteins such as intracellular accumulation of α-Syn (detected as aggregates of pS129-α-Syn) and phosphorylation of LRRK2 kinase at residue S935. In addition, mutant DALNs showed a 17.16- and 6.17-fold decrease in DA-induced Ca2+ flux, compared to WT DALNs. These observations suggest that iAβ and p-TAU, together with p-α-Syn, and p-LRRK2 kinase, may damage DAergic neurons and thereby contribute to the exacerbation of neuropathologic processes in FAD. Remarkably, the LRRK2 inhibitor PF-06447475 (PF-475) significantly reversed PSEN1 I416T-induced neuropathological markers in DAergic neurons. PF-465 inhibitor reduced iAβ, oxDJ-1C106-SO3, and p-TAU. In addition, this inhibitor reduced pS935-LRRK2, pS129-αSYN, pS63-c-JUN, and CC3. We conclude that the observed neuroprotective effects of PF-475 are due to direct inhibition of LRRK2 activity and that the LRRK2 protein is upstream of the molecular cascade of apoptosis and proteinopathy. Our results suggest that PF-475 is an effective neuroprotective agent against endogenous PSEN1 I416T-induced neurotoxicity in DALNs coexisting with Parkinson's disease markers. Therefore, PF-475 may be of great therapeutic value in FAD.
Collapse
Affiliation(s)
| | | | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Institute of Medical Research, Faculty of Medicine, University Research Headquarters, Calle 62#52-59, Building 1, Laboratory 411/412, Medellin 050010, Colombia; (D.A.Q.-E.); (C.V.-P.)
| |
Collapse
|
6
|
Al-Wahaibi LH, Elshamsy AM, Ali TFS, Youssif BGM, Bräse S, Abdel-Aziz M, El-Koussi NA. Design, synthesis, in silico studies, and apoptotic antiproliferative activity of novel thiazole-2-acetamide derivatives as tubulin polymerization inhibitors. Front Chem 2025; 13:1565699. [PMID: 40308265 PMCID: PMC12040969 DOI: 10.3389/fchem.2025.1565699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction Tubulin polymerization inhibitors have emerged as interesting anticancer therapies. We present the design, synthesis, and structural elucidation of novel thiazole-based derivatives to identify novel tubulin inhibitors with potent antiproliferative efficacy and strong inhibition of tubulin polymerization. Methods The novel compounds consist of two scaffolds. Scaffold A compounds 10a-e and scaffold B compounds 13a-e. the structures of the newly synthesized compounds 10a-e and 13a-e were validated using 1H NMR, 13C NMR, and elemental analysis. Results and Discussion The most effective antitubulin derivative was 10a, exhibiting an IC50 value of 2.69 μM. Subsequently, 10o and 13d exhibited IC50 values of 3.62 μM and 3.68 μM, respectively. These compounds exhibited more potency than the reference combretastatin A-4, which displayed an IC50 value of 8.33 μM. These compounds had no cytotoxic effects on normal cells, preserving over 85% cell viability at 50 μM. The antiproliferative experiment demonstrated that compounds 10a, 10o, and 13d displayed significant activity against four cancer cell lines, with average GI50 values of 6, 7, and 8 μM, equivalent to the reference's doxorubicin and sorafenib. Compounds 10a, 10o, and 13d were demonstrated to activate caspases 3, 9, and Bax, while down-regulating the anti-apoptotic protein Bcl2. Molecular docking studies demonstrated superior binding affinities for 10a (-7.3 kcal/mol) at the colchicine binding site of tubulin, forming key hydrophobic and hydrogen bonding interactions that enhance its activity. ADMET analysis confirmed favorable drug-like properties, establishing these compounds as promising candidates for further development as anticancer agents targeting tubulin polymerization.
Collapse
Affiliation(s)
- Lamya H. Al-Wahaibi
- Department of Chemistry, College of Sciences, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ali M. Elshamsy
- Pharmceutical Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Taha F. S. Ali
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Bahaa G. M. Youssif
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Nawal A. El-Koussi
- Pharmceutical Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
7
|
Fabiano AR, Newman MW, Dombroski JA, Rowland SJ, Knoblauch SV, Kusunose J, Gibson‐Corley KN, Kaufman BG, Ren L, Caskey CF, King MR. Applying Ultrasound to Mechanically and Noninvasively Sensitize Prostate Tumors to TRAIL-Mediated Apoptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412995. [PMID: 39976192 PMCID: PMC12005757 DOI: 10.1002/advs.202412995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/26/2025] [Indexed: 02/21/2025]
Abstract
Non-surgical and safe prostate cancer (PCa) therapies are in demand. Soluble tumor necrosis factor (TNF-α) related apoptosis inducing ligand (TRAIL), a cancer-specific drug, shows preclinical efficacy but has a short circulation half-life. This research has shown that physiological fluid shear stress activates mechanosensitive ion channels (MSCs), such as Piezo1, enhancing TRAIL-mediated apoptosis in cancer cells. Herein, noninvasive, focal ultrasound (FUS) is implemented to augment the pro-apoptotic effects of TRAIL. Using thermally safe FUS parameters, it is observed that TRAIL sensitivity increases with higher FUS pressure in PCa cells, mediated by Piezo1. This is confirmed by examining the effects of calcium chelation, MSC inhibitors, and PIEZO knockdown. In vivo, a multi-dose study with 10 min FUS exposure shows that 0 and 4-h intervals between TRAIL and FUS significantly reduce tumor burden, with an increase in apoptosis evident by enhanced cleaved-caspase 3 expression. This mechanotherapy offers a clinically translatable approach by utilizing widely available FUS technology, applicable to treat additional cancer types.
Collapse
Affiliation(s)
- Abigail R. Fabiano
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
- Department of BioengineeringRice UniversityHoustonTX77005USA
| | - Malachy W. Newman
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Jenna A. Dombroski
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Schyler J. Rowland
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | | | - Jiro Kusunose
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTN37235USA
| | - Katherine N. Gibson‐Corley
- Department of PathologyMicrobiology and ImmunologyDivision of Comparative MedicineVanderbilt University Medical CenterNashvilleTN37235USA
| | | | - Liqin Ren
- Department of BioengineeringRice UniversityHoustonTX77005USA
| | - Charles F. Caskey
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTN37235USA
- Department of Radiology and Radiological SciencesVanderbilt UniversityNashvilleTN37235USA
| | - Michael R. King
- Department of BioengineeringRice UniversityHoustonTX77005USA
| |
Collapse
|
8
|
Li Q, Chang X, Han Y, Guo Z, Liu Y, Guo B, Liu C, Yang B, Fan Z, Jiang H, Chang X. Consumption of Endogenous Caspase-3 Activates Molecular Theranostic Nanoplatform against Inflammation-Induced Profibrotic Positive Feedback in Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412303. [PMID: 39686776 PMCID: PMC11809389 DOI: 10.1002/advs.202412303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/30/2024] [Indexed: 12/18/2024]
Abstract
The limited and backward diagnostic approaches elicit high mortality associated with pulmonary fibrosis (PF) because they fail to identify injury phase of PF. Developing a precisely theranostic nanoplatform presents a promising shortcut to reverse PF. Herein, a specific molecular nanotheranostic (Casp-GNMT), which is triggered by endogenous cysteinyl aspartate specific proteinase-3 (caspase-3), boosts antifibrotic efficacy through bioimaging synergistic with chemotherapy at molecular level, facilitating by ionizable lipid and reactive oxygen species sensitive lipid for precise and manageable therapy. The activation of molecular imaging probe (pCY-pairs) by consumption of endogenous caspase-3 initiates fluorescence resonance energy transfer-guided theranostic pattern, aiming to restore mitochondrial dysfunction-induced oxidative stress and inflammatory responses in alveolar epithelial cells II (AECs II). This process sequentially resists the expression of interleukin-1β and vascular endothelial growth factor receptor through combined with nintedanib, further suppressing abnormal injury of AECs II and persistent migration and proliferation of inflammatory cells. Especially, the homeostasis of injured AECs II diminishes excessive accumulation of transforming growth factor-β to restrain myofibroblasts proliferation and collagen deposition, thereby amplifying the possibility of reversing PF. This theranostic nanoplatform is proposed to provide a prompt and exact approach to enhance diagnostic authenticity and treating efficiency through harnessing endogenous indicator for PF reversal.
Collapse
Affiliation(s)
- Qiu‐Ling Li
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Xin Chang
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
- Liaoning Provincial Key Laboratory of Marine Bioactive SubstancesJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Yu‐Mo Han
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Zi‐Chao Guo
- The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Yi‐Na Liu
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Bin Guo
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
- Liaoning Provincial Key Laboratory of Marine Bioactive SubstancesJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Chang Liu
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Bin‐Rong Yang
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
- Liaoning Provincial Key Laboratory of Marine Bioactive SubstancesJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Zhong‐Kai Fan
- The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Hu‐Lin Jiang
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
- Jiangsu Key Laboratory of Druggability of BiopharmaceuticalsChina Pharmaceutical UniversityNanjingJiangsu210009China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and ExcipientsChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Xin Chang
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
- Liaoning Provincial Key Laboratory of Marine Bioactive SubstancesJinzhou Medical UniversityJinzhouLiaoning121001China
| |
Collapse
|
9
|
Samarasekera G, Go NE, Choutka C, Xu J, Takemon Y, Chan J, Chan M, Perera S, Aparicio S, Morin GB, Marra MA, Chittaranjan S, Gorski SM. Caspase 3 and caspase 7 promote cytoprotective autophagy and the DNA damage response during non-lethal stress conditions in human breast cancer cells. PLoS Biol 2025; 23:e3003034. [PMID: 39982959 PMCID: PMC11882052 DOI: 10.1371/journal.pbio.3003034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/05/2025] [Accepted: 01/24/2025] [Indexed: 02/23/2025] Open
Abstract
Cell stress adaptation plays a key role in normal development and in various diseases including cancer. Caspases are activated in response to cell stress, and growing evidence supports their function in non-apoptotic cellular processes. A role for effector caspases in promoting stress-induced cytoprotective autophagy was demonstrated in Drosophila, but has not been explored in the context of human cells. We found a functionally conserved role for effector caspase 3 (CASP3) and caspase 7 (CASP7) in promoting starvation or proteasome inhibition-induced cytoprotective autophagy in human breast cancer cells. The loss of CASP3 and CASP7 resulted in an increase in PARP1 cleavage, reduction in LC3B and ATG7 transcript levels, and a reduction in H2AX phosphorylation, consistent with a block in autophagy and DNA damage-induced stress response pathways. Surprisingly, in non-lethal cell stress conditions, CASP7 underwent non-canonical processing at two calpain cleavage sites flanking a PARP1 exosite, resulting in stable CASP7-p29/p30 fragments. Expression of CASP7-p29/p30 fragment(s) could rescue H2AX phosphorylation in the CASP3 and CASP7 double knockout background. Strikingly, yet consistent with these phenotypes, the loss of CASP3 and CASP7 exhibited synthetic lethality with BRCA1 loss. These findings support a role for human caspases in stress adaptation through PARP1 modulation and reveal new therapeutic avenues for investigation.
Collapse
Affiliation(s)
- Gayathri Samarasekera
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nancy E. Go
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Courtney Choutka
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Jing Xu
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Yuka Takemon
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Genome Science and Technology Graduate Program, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jennifer Chan
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Michelle Chan
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Shivani Perera
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Samuel Aparicio
- Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gregg B. Morin
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marco A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Suganthi Chittaranjan
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Sharon M. Gorski
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
10
|
Zhu C, Whitcomb LA, Chicco AJ, Gravely ME, Alcocer HM, Alambarrio DA, Gonzalez JM, Smith CL, Nair MN, Loh HY, Engle TE, Niraula A, Zhai C. Effects of Nicotinamide Riboside Supplementation on Postmortem Mitochondrial Functionality and Apoptotic Activation. Metabolites 2025; 15:31. [PMID: 39852374 PMCID: PMC11766954 DOI: 10.3390/metabo15010031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Early postmortem mitochondrial function and apoptotic activation affect meat quality development. Nicotinamide riboside (NR) supplementation to pigs prior to harvest can improve pork color stability, but its mechanism remains unclear. This study aimed to evaluate the impact of NR supplementation on early postmortem mitochondrial functionality and apoptosis. METHODS Sixteen pigs (N = 16) were individually fed a control or NR-supplemented diet (30 mg·kg body weight-1·d-1) for 10 days prior to harvest. Longissimus dorsi muscle samples were collected at 45 min and 24 h postmortem and analyzed for mitochondrial functionality using high-resolution respirometry and apoptotic protein abundance (apoptosis regulator Bcl-2-associated X (BAX), apoptotic inducing factor (AIF), and caspase 3 (CASP3)) via immunoblotting. RESULTS NR-supplemented muscle exhibited lower proton leak-associated respiration at 45 min postmortem (p < 0.05), followed by a slower accumulation of mitochondrial outer membrane permeabilization (MOMP; p < 0.05) and a slower loss of mitochondrial integral function (p < 0.05) from 45 min to 24 h postmortem. NR supplementation decreased BAX abundance at 45 min postmortem but increased mature AIF abundance (62 kDa) at 24 h postmortem (p < 0.05). The abundance of CASP3 fragments (~29 kDa) decreased from 45 min to 24 h postmortem, independent of treatment (p < 0.05). CONCLUSIONS NR supplementation demonstrated the potential to protect mitochondrial integral function and alleviate apoptotic activation in early postmortem porcine skeletal muscle, which might contribute to a higher meat color stability in NR-supplemented pork during retail display.
Collapse
Affiliation(s)
- Chen Zhu
- Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA; (C.Z.); (A.N.)
| | - Luke A. Whitcomb
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (L.A.W.); (A.J.C.)
| | - Adam J. Chicco
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (L.A.W.); (A.J.C.)
| | - Morgan E. Gravely
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA; (M.E.G.); (H.M.A.); (D.A.A.); (J.M.G.)
| | - Hanna M. Alcocer
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA; (M.E.G.); (H.M.A.); (D.A.A.); (J.M.G.)
| | - Daniela A. Alambarrio
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA; (M.E.G.); (H.M.A.); (D.A.A.); (J.M.G.)
| | - John M. Gonzalez
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA; (M.E.G.); (H.M.A.); (D.A.A.); (J.M.G.)
| | - Colton L. Smith
- Department of Animal Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.L.S.); (M.N.N.); (H.Y.L.); (T.E.E.)
| | - Mahesh N. Nair
- Department of Animal Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.L.S.); (M.N.N.); (H.Y.L.); (T.E.E.)
| | - Huey Yi Loh
- Department of Animal Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.L.S.); (M.N.N.); (H.Y.L.); (T.E.E.)
| | - Terry E. Engle
- Department of Animal Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.L.S.); (M.N.N.); (H.Y.L.); (T.E.E.)
| | - Arya Niraula
- Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA; (C.Z.); (A.N.)
| | - Chaoyu Zhai
- Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA; (C.Z.); (A.N.)
| |
Collapse
|
11
|
Mohamed AM, Abou-Ghadir OMF, Mostafa YA, Almarhoon ZM, Bräse S, Youssif BGM. Design, synthesis, and antiproliferative activity of new 1,2,3-triazole/quinazoline-4-one hybrids as dual EGFR/BRAF V600E inhibitors. RSC Adv 2024; 14:38403-38415. [PMID: 39640522 PMCID: PMC11618052 DOI: 10.1039/d4ra06694d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
A novel series of 1,2,3-triazole/quinazoline-4-one hybrids (8a-t) were designed and synthesized as dual-targeted antiproliferative agents. Compounds 8a-t were evaluated for their antiproliferative efficacy against a panel of four cancer cell lines. The results indicated that most of the evaluated compounds exhibited strong antiproliferative activity, with 8f, 8g, 8h, 8j, and 8l demonstrating the highest potency. These five compounds were investigated as EGFR and BRAFV600E inhibitors. The in vitro tests showed that compounds 8g, 8h, and 8j are strong antiproliferative agents that might work as dual EGFR/BRAFV600E inhibitors. Compounds 8g and 8h were further examined as activators of caspases 3, 8, and Bax and down-regulators of the anti-apoptotic protein Bcl2. The results indicated that the studied compounds had considerable apoptotic antiproliferative action. The investigation of the cell cycle and apoptosis revealed that compound 8g induces cell cycle arrest during the G1 phase transition. Molecular docking experiments are thoroughly examined to validate the binding interactions of the most active hybrids with the active sites of EGFR and BRAFV600E. The data indicated that the examined compounds can efficiently engage with essential amino acid residues in both kinases.
Collapse
Affiliation(s)
- Amira M Mohamed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt
| | - Ola M F Abou-Ghadir
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt
| | - Yaser A Mostafa
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt
| | - Zainab M Almarhoon
- Department of Chemistry, College of Science, King Saud University Riyadh 11451 Saudi Arabia
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology 76131 Karlsruhe Germany
| | - Bahaa G M Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt
| |
Collapse
|
12
|
You YL, Byun HJ, Lee JS, Choi HS, Youk JS. Euonymus alatus and its compounds suppress hydrogen peroxide-induced oxidative stress in HT22 cells. Food Sci Biotechnol 2024; 33:3567-3577. [PMID: 39493395 PMCID: PMC11525359 DOI: 10.1007/s10068-024-01601-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 11/05/2024] Open
Abstract
This study aimed to explore the protective effects of Euonymus alatus (EA) leaves and its compounds on hydrogen peroxide (H2O2)-induced neuronal cell death. EA effectively reversed the H2O2-induced decrease in HT22 cell viability. Anti-apoptotic marker poly(ADP-ribose) polymerase significantly increased with EA treatment, whereas BAX/BCL2 and cleaved caspase-3/procaspase-3 ratios, which represent apoptotic markers, were dose-dependently decreased by EA treatment. Additionally, EA effectively decreased β-secretase production, acetylcholine esterase activity, and Tau phosphorylation, pathological features observed in Alzheimer's disease. Furthermore, EA significantly increased the protein levels of NRF2 and HO-1, as well as the gene expression of antioxidant enzymes, including catalase, superoxide dismutase 1, and glutathione peroxidase. LC-MS/MS and HPLC analyses revealed the presence of chlorogenic acid and leucosides in EA. Both chlorogenic acid and leucosides showed protective effects against H2O2-induced neuronal cell death. This study highlights the potential of EA and its compounds as functional edible agents for neuroprotection against oxidative stress. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-024-01601-4.
Collapse
Affiliation(s)
- Ye-Lim You
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-gil 20, Jongno-gu, Seoul, 03016 Republic of Korea
| | - Ha-Jun Byun
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-gil 20, Jongno-gu, Seoul, 03016 Republic of Korea
| | - Jeong Soon Lee
- Forest Environment Research Institute of Gyeongsangbuk-do, Gyeongju, 38174 Republic of Korea
| | - Hyeon-Son Choi
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-gil 20, Jongno-gu, Seoul, 03016 Republic of Korea
| | - Jin-Soo Youk
- Department of Food & Beverage Management, Hanyang Women’s University, 200, Salgoji-gil, Seongdong-gu, Seoul, Republic of Korea
| |
Collapse
|
13
|
Yousef RG, Eissa IH, Elkady H, M Mehany AB, Abo-Saif MA, Radwan MM, ElSohly MA, Ibrahim IM, Elwan A, El-Zahabi MA. Design and synthesis of new nicotinamides as immunomodulatory VEGFR-2 inhibitors and apoptosis inducers. Future Med Chem 2024; 16:2583-2598. [PMID: 39540547 PMCID: PMC11731296 DOI: 10.1080/17568919.2024.2421150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Nicotinamide-based VEGFR-2 inhibitors have good contribution in drug discovery.Aim: Development of novel nicotinamides as VEGFR-2 inhibitors.Methods: different in vitro and in silico assays were conducted to evaluate the VEGFR-2 inhibition and cytotoxicity.Results: Compound 16c displayed strongest anti-VEGFR-2 potentiality and good anti-proliferative effects. Compound 16c enhanced apoptosis and caused cell cycle arrest in the Pre-G1 and S phases. Compound 16c boosted the level of the apoptotic caspase-3 and inhibited the level of TNF-α and IL-6 in tumor cells. Molecular docking and molecular dynamics (MD) simulations indicated the outstanding binding potential of compound 16c against VEGFR-2.Conclusion: Compound 16c is a good candidate for the creation of a novel antiangiogenic lead anticancer medication.
Collapse
Affiliation(s)
- Reda G Yousef
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Hazem Elkady
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Ahmed B M Mehany
- Zoology Department, Faculty of Science (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Mariam Ali Abo-Saif
- Biochemistry Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Mohamed M Radwan
- National Center for Natural Products Research, University of Mississippi, University, MS38677, USA
| | - Mahmoud A ElSohly
- National Center for Natural Products Research, University of Mississippi, University, MS38677, USA
| | - Ibrahim M Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Alaa Elwan
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Mohamed Ayman El-Zahabi
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| |
Collapse
|
14
|
Batista Brochado AC, de Moraes JA, Rodrigues de Oliveira B, De Souza Lima VH, Mariano ED, Karande S, Romasco T, Leite PEC, Mourão CF, Gomes Alves G. Metabolic and Regulatory Pathways Involved in the Anticancer Activity of Perillyl Alcohol: A Scoping Review of In Vitro Studies. Cancers (Basel) 2024; 16:4003. [PMID: 39682189 PMCID: PMC11640718 DOI: 10.3390/cancers16234003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Perillyl alcohol (POH), a plant-derived compound, has demonstrated anti-tumor activity across various human cancers. Understanding the regulatory pathways through which POH exerts its effects is crucial for identifying new therapeutic opportunities and exploring potential drug repositioning strategies. Therefore, this scoping review aims to provide a comprehensive overview of the metabolic and regulatory pathways involved in the anticancer effects of POH, based on in vitro evidence. METHODS Following the PRISMA-ScR 2018 guidelines, a systematic search was conducted in the PUBMED, Web of Science, and Scopus databases. RESULTS A total of 39 studies were included, revealing that POH exerts its biological effects by modulating several pathways, including the regulation of cyclins, CDKs, and p21, thereby affecting cell cycle progression. It inhibits growth and promotes cell death by attenuating AKT phosphorylation, reducing PARP-1 activity, increasing caspase activity and the FAS receptor and its ligand FASL. Additionally, POH reduces ERK phosphorylation, inhibits RAS protein isoprenylation, and decreases Na/K-ATPase activity. CONCLUSIONS In conclusion, this review delineates the key regulatory pathways responsible for mediating the biological effects of POH in cancer.
Collapse
Affiliation(s)
- Ana Carolina Batista Brochado
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
| | - Júlia Alves de Moraes
- Clinical Research Unit, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi 24020-140, Brazil
| | - Bruna Rodrigues de Oliveira
- Clinical Research Unit, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi 24020-140, Brazil
| | - Victor Hugo De Souza Lima
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
| | | | - Sachin Karande
- Dental Research Division, Department of Periodontology and Oral Implantology, Fluminense Federal University, Niteroi 21941-617, Brazil
| | - Tea Romasco
- Division of Dental Research Administration, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Paulo Emilio Correa Leite
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
| | - Carlos Fernando Mourão
- Department of Basic and Clinical Translational Sciences, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Gutemberg Gomes Alves
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
- Clinical Research Unit, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi 24020-140, Brazil
| |
Collapse
|
15
|
Kalantari L, Hajjafari A, Goleij P, Rezaee A, Amirlou P, Farsad S, Foroozand H, Arefnezhad R, Rezaei-Tazangi F, Jahani S, Yazdani T, Nazari A. Umbilical cord mesenchymal stem cells: A powerful fighter against colon cancer? Tissue Cell 2024; 90:102523. [PMID: 39154502 DOI: 10.1016/j.tice.2024.102523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
Colon cancer (CC) stands as one of the most common malignancies related to the gastrointestinal system, whose increasing incidence and death rates have been reported all over the world. Standard treatments for fighting cancers like CC comprise surgical approaches, chemotherapy, and radiotherapy, which are suggested by clinicians according to patients' conditions and disease stages. However, patients who utilize these modalities may suffer from serious side effects and adverse outcomes, for example, toxicity and tumor recurrence, as well as a low 5-year survival rate. The present shreds of evidence showed that mesenchymal stem cells (MSCs) can have a suitable capacity for treating different health problems, especially neoplasms. These multipotent stem cells can be isolated from several sources, such as the umbilical cord, bone marrow, adipose tissue, and placenta. Among these mesenchymal sources, umbilical cord-MSCs have gathered much attention in scientific societies due to their advantages (e.g., low immunogenicity, lack of ethical problems, and easy collection). These days, the efficacy of umbilical cord-MSCs and umbilical cord-MSCs-based strategies, such as conditioned medium, extracellular vesicles, and exosomes, on CC have been explored, and promising findings have been stated. Therefore, in this review, we aimed to summarize and debate evidence regarding the effects of UC-MSCs and their related products on CC with a focus on molecular and cellular mechanisms involved in its treatment and pathogenesis of this malignant tumor.
Collapse
Affiliation(s)
- Leila Kalantari
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran; School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ashkan Hajjafari
- Department of Pathobiology, Faculty of Veterinary Medicine Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Pouya Goleij
- Department of Genetics, Sana Institute of Higher Education, Sari, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parsa Amirlou
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shirin Farsad
- Faculty of Basic Science, Islamic Azad University, Qom, Iran
| | - Hassan Foroozand
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Arefnezhad
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Coenzyme R Research Institute, Tehran, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Saleheh Jahani
- Pathology department, University of California, SanDiego, United States
| | - Taha Yazdani
- Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Nazari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Ge J, Wang Y, Li X, Lu Q, Yu H, Liu H, Ma K, Deng X, Luo ZQ, Liu X, Qiu J. Phosphorylation of caspases by a bacterial kinase inhibits host programmed cell death. Nat Commun 2024; 15:8464. [PMID: 39349471 PMCID: PMC11442631 DOI: 10.1038/s41467-024-52817-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/20/2024] [Indexed: 10/02/2024] Open
Abstract
The intracellular bacterial pathogen Legionella pneumophila utilizes the Dot/Icm system to translocate over 330 effectors into the host cytosol. These virulence factors modify a variety of cell processes, including pathways involved in cell death and survival, to promote bacterial proliferation. Here, we show that the effector LegK3 is a eukaryotic-like Ser/Thr kinase that functions to suppress host apoptosis. Mechanistically, LegK3 directly phosphorylates multiple caspases involved in apoptosis signaling, including Caspase-3, Caspase-7, and Caspase-9. LegK3-induced phosphorylation of these caspases occurs at serine (Ser29 in Caspase-3 and Ser199 in Caspase-7) or threonine (Thr102 in Caspase-9) residues located in the prodomain or interdomain linkers. These modifications interfere with the suitability of the caspases as the substrates of initiator caspases or upstream regulators without impacting their proteolytic activity. Collectively, our study reveals a novel strategy used by L. pneumophila to maintain the integrity of infected cells for its intracellular growth.
Collapse
Affiliation(s)
- Jinli Ge
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ying Wang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Xueyu Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qian Lu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hangqian Yu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hongtao Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Kelong Ma
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuming Deng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhao-Qing Luo
- Purdue Institute for Inflammation, Immunology and Infectious Disease and Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Xiaoyun Liu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China.
| | - Jiazhang Qiu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
17
|
Moreira S, Martins AD, Alves MG, Pastor LM, Seco-Rovira V, Oliveira PF, Pereira MDL. Aminocarb Exposure Induces Cytotoxicity and Endoplasmic Reticulum Stress-Mediated Apoptosis in Mouse Sustentacular Sertoli Cells: Implications for Male Infertility and Environmental Health. BIOLOGY 2024; 13:721. [PMID: 39336148 PMCID: PMC11429014 DOI: 10.3390/biology13090721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Exposure to pesticides, poses a significant threat to male fertility by compromising crucial cells involved in spermatogenesis. Aminocarb, is a widely used carbamate insecticide, although its detrimental effects on the male reproductive system, especially on sustentacular Sertoli cells, pivotal for spermatogenesis, remains poorly understood. In this study, we investigated the effects of escalating concentrations of aminocarb on a mouse Sertoli cell line, TM4. Assessments included cytotoxic analysis, mitochondrial biogenesis and membrane potential, expression of apoptotic proteins, caspase-3 activity, and oxidative stress evaluation. Our findings revealed a dose-dependent reduction in the proliferation and viability of TM4 cells following exposure to increasing concentrations of aminocarb. Notably, exposure to 5 μM of aminocarb induced depolarization of mitochondria membrane potential, and a significant decrease in the ratio of phosphorylated eIF2α to total eIF2α, suggesting heightened endoplasmic reticulum stress via the activation of the eIF2α pathway. Moreover, the same aminocarb concentration was demonstrated to increase both caspase-3 protein levels and activity, indicating an apoptotic induction. Collectively, our results demonstrate that aminocarb serves as an apoptotic inducer for mouse sustentacular Sertoli cells in vitro, suggesting its potential to modulate independent pathways of the apoptotic cascade. These findings underscore the deleterious impact of aminocarb on spermatogenic performance and male fertility, highlighting the urgent need for further investigation into its mechanisms of action and mitigation strategies to safeguard male fertility.
Collapse
Affiliation(s)
- Sílvia Moreira
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- CICECO-Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana D Martins
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Marco G Alves
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Luis Miguel Pastor
- Departamento de Biología Celular e Histología, Faculdad de Medicina, IMIB-Arrixaca, Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia, 30120 Murcia, Spain
| | - Vicente Seco-Rovira
- Departamento de Biología Celular e Histología, Faculdad de Medicina, IMIB-Arrixaca, Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia, 30120 Murcia, Spain
| | - Pedro F Oliveira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Maria de Lourdes Pereira
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- CICECO-Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
18
|
Kumar A, Angelopoulou E, Pyrgelis ES, Piperi C, Mishra A. Harnessing Therapeutic Potentials of Biochanin A in Neurological Disorders: Pharmacokinetic and Pharmacodynamic Overview. Chem Biodivers 2024; 21:e202400709. [PMID: 38828832 DOI: 10.1002/cbdv.202400709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/05/2024]
Abstract
Biochanin A, an isoflavone flavonoid with estrogenic activity, is naturally found in red clover and other legumes. It possesses a wide range of pharmacological properties, including antioxidant, anti-inflammatory, anti-apoptotic, neuroprotective, and anticancer effects. In recent years, a growing body of pre-clinical research has focused on exploring the therapeutic potential of biochanin A in various neurological disorders, such as Alzheimer's and Parkinson's disease, multiple sclerosis, epilepsy, ischemic brain injury, gliomas, and neurotoxicity. This comprehensive review aims to shed light on the underlying molecular mechanisms that contribute to the neuroprotective role of biochanin A based on previous pre-clinical studies. Furthermore, it provides a detailed overview of the protective effects of biochanin A in diverse neurological disorders. The review also addresses the limitations associated with biochanin A administration and discusses different approaches employed to overcome these challenges. Finally, it highlights the future opportunities for translating biochanin A from pre-clinical research to clinical studies while also considering its commercial viability as a dietary supplement or a potential treatment for various diseases.
Collapse
Affiliation(s)
- Ankit Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Guwahati, Changsari, Kamrup, Assam, 781101, India
| | - Efthalia Angelopoulou
- Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528, Athens, Greece
| | - Efstratios-Stylianos Pyrgelis
- Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Awanish Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Guwahati, Changsari, Kamrup, Assam, 781101, India
| |
Collapse
|
19
|
Aguilar EN, Sagar S, Murray BR, Rajesh C, Lei EK, Michaud SA, Goodlett DR, Caffrey TC, Grandgenett PM, Swanson B, Brooks TM, Black AR, van Faassen H, Hussack G, Henry KA, Hollingsworth MA, Brooks CL, Radhakrishnan P. Structural Basis for Multivalent MUC16 Recognition and Robust Anti-Pancreatic Cancer Activity of Humanized Antibody AR9.6. Mol Cancer Ther 2024; 23:836-853. [PMID: 38394685 PMCID: PMC11660185 DOI: 10.1158/1535-7163.mct-23-0868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/25/2024] [Accepted: 02/21/2024] [Indexed: 02/25/2024]
Abstract
Mucin-16 (MUC16) is a target for antibody-mediated immunotherapy in pancreatic ductal adenocarcinoma (PDAC) among other malignancies. The MUC16-specific monoclonal antibody AR9.6 has shown promise for PDAC immunotherapy and imaging. Here, we report the structural and biological characterization of the humanized AR9.6 antibody (huAR9.6). The structure of huAR9.6 was determined in complex with a MUC16 SEA (Sea urchin sperm, Enterokinase, Agrin) domain. Binding of huAR9.6 to recombinant, shed, and cell-surface MUC16 was characterized, and anti-PDAC activity was evaluated in vitro and in vivo. HuAR9.6 bound a discontinuous, SEA domain epitope with an overall affinity of 88 nmol/L. Binding affinity depended on the specific SEA domain(s) present, and glycosylation modestly enhanced affinity driven by favorable entropy and enthalpy and via distinct transition state thermodynamic pathways. Treatment with huAR9.6 reduced the in vitro growth, migration, invasion, and clonogenicity of MUC16-positive PDAC cells and patient-derived organoids (PDO). HuAR9.6 blocked MUC16-mediated ErbB and AKT activation in PDAC cells, PDOs, and patient-derived xenografts and induced antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. More importantly, huAR9.6 treatment caused substantial PDAC regression in subcutaneous and orthotopic tumor models. The mechanism of action of huAR9.6 may depend on dense avid binding to homologous SEA domains on MUC16. The results of this study validate the translational therapeutic potential of huAR9.6 against MUC16-positive PDACs.
Collapse
Affiliation(s)
- Eric N. Aguilar
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA, USA
| | - Satish Sagar
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brandy R. Murray
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA, USA
| | - Christabelle Rajesh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric K. Lei
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Sarah A. Michaud
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - David R. Goodlett
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Thomas C. Caffrey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paul M. Grandgenett
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benjamin Swanson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Teresa M. Brooks
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA, USA
| | - Adrian R. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Henk van Faassen
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Greg Hussack
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Kevin A. Henry
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cory L. Brooks
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
20
|
Myoli A, Choene M, Kappo AP, Madala NE, van der Hooft JJJ, Tugizimana F. Charting the Cannabis plant chemical space with computational metabolomics. Metabolomics 2024; 20:62. [PMID: 38796627 PMCID: PMC11127828 DOI: 10.1007/s11306-024-02125-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/02/2024] [Indexed: 05/28/2024]
Abstract
INTRODUCTION The chemical classification of Cannabis is typically confined to the cannabinoid content, whilst Cannabis encompasses diverse chemical classes that vary in abundance among all its varieties. Hence, neglecting other chemical classes within Cannabis strains results in a restricted and biased comprehension of elements that may contribute to chemical intricacy and the resultant medicinal qualities of the plant. OBJECTIVES Thus, herein, we report a computational metabolomics study to elucidate the Cannabis metabolic map beyond the cannabinoids. METHODS Mass spectrometry-based computational tools were used to mine and evaluate the methanolic leaf and flower extracts of two Cannabis cultivars: Amnesia haze (AMNH) and Royal dutch cheese (RDC). RESULTS The results revealed the presence of different chemical compound classes including cannabinoids, but extending it to flavonoids and phospholipids at varying distributions across the cultivar plant tissues, where the phenylpropnoid superclass was more abundant in the leaves than in the flowers. Therefore, the two cultivars were differentiated based on the overall chemical content of their plant tissues where AMNH was observed to be more dominant in the flavonoid content while RDC was more dominant in the lipid-like molecules. Additionally, in silico molecular docking studies in combination with biological assay studies indicated the potentially differing anti-cancer properties of the two cultivars resulting from the elucidated chemical profiles. CONCLUSION These findings highlight distinctive chemical profiles beyond cannabinoids in Cannabis strains. This novel mapping of the metabolomic landscape of Cannabis provides actionable insights into plant biochemistry and justifies selecting certain varieties for medicinal use.
Collapse
Affiliation(s)
- Akhona Myoli
- Department of Biochemistry, University of Johannesburg, Auckland Park, Johannesburg, 2006, South Africa
| | - Mpho Choene
- Department of Biochemistry, University of Johannesburg, Auckland Park, Johannesburg, 2006, South Africa
| | - Abidemi Paul Kappo
- Department of Biochemistry, University of Johannesburg, Auckland Park, Johannesburg, 2006, South Africa
| | | | - Justin J J van der Hooft
- Department of Biochemistry, University of Johannesburg, Auckland Park, Johannesburg, 2006, South Africa.
- Bioinformatics Group, Wageningen University, Wageningen, 6708 PB, the Netherlands.
| | - Fidele Tugizimana
- Department of Biochemistry, University of Johannesburg, Auckland Park, Johannesburg, 2006, South Africa.
- International Research and Development Division, Omnia Group, Ltd., Bryanston, Johannesburg, 2021, South Africa.
- National Institute for Theoretical and Computational Sciences, Johannesburg, South Africa.
| |
Collapse
|
21
|
Tu TH, Grunbaum A, Santinon F, Kazanova A, Rozza N, Kremer R, Mihalcioiu C, Rudd CE. Decreased progenitor TCF1 + T-cells correlate with COVID-19 disease severity. Commun Biol 2024; 7:526. [PMID: 38702425 PMCID: PMC11068881 DOI: 10.1038/s42003-024-05922-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/16/2024] [Indexed: 05/06/2024] Open
Abstract
COVID-19, caused by SARS-CoV-2, can lead to a severe inflammatory disease characterized by significant lymphopenia. However, the underlying cause for the depletion of T-cells in COVID-19 patients remains incompletely understood. In this study, we assessed the presence of different T-cell subsets in the progression of COVID-19 from mild to severe disease, with a focus on TCF1 expressing progenitor T-cells that are needed to replenish peripheral T-cells during infection. Our results showed a preferential decline in TCF1+ progenitor CD4 and CD8+ T-cells with disease severity. This decline was seen in various TCF1+ subsets including naive, memory and effector-memory cells, and surprisingly, was accompanied by a loss in cell division as seen by a marked decline in Ki67 expression. In addition, TCF1+ T-cells showed a reduction in pro-survival regulator, BcL2, and the appearance of a new population of TCF1 negative caspase-3 expressing cells in peripheral blood from patients with severe disease. The decline in TCF1+ T-cells was also seen in a subgroup of severe patients with vitamin D deficiency. Lastly, we found that sera from severe patients inhibited TCF1 transcription ex vivo which was attenuated by a blocking antibody against the cytokine, interleukin-12 (IL12). Collectively, our findings underscore the potential significance of TCF1+ progenitor T-cells in accounting for the loss of immunity in severe COVID-19 and outline an array of markers that could be used to identify disease progression.
Collapse
Affiliation(s)
- Thai Hien Tu
- Départment of Medicine, Universite de Montreal, Montreal, QC, H3T 1J4, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Division of Immunology-Oncology, Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, H1T 2M4, Canada
| | - Ami Grunbaum
- Division of Experimental Medicine, McGill University, Montreal, QC, H3A 0G4, Canada
- Department of Medicine, Research Institute of the McGill University Health Center, Montreal, H3A 0G4, Canada
- Division of Medical Biochemistry, McGill University Health Centre, Montréal, QC, Canada
| | - François Santinon
- Départment of Medicine, Universite de Montreal, Montreal, QC, H3T 1J4, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Division of Immunology-Oncology, Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, H1T 2M4, Canada
| | - Alexandra Kazanova
- Départment of Medicine, Universite de Montreal, Montreal, QC, H3T 1J4, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Division of Immunology-Oncology, Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, H1T 2M4, Canada
| | - Nicholas Rozza
- Division of Experimental Medicine, McGill University, Montreal, QC, H3A 0G4, Canada
- Department of Medicine, Research Institute of the McGill University Health Center, Montreal, H3A 0G4, Canada
| | - Richard Kremer
- Division of Experimental Medicine, McGill University, Montreal, QC, H3A 0G4, Canada
- Department of Medicine, Research Institute of the McGill University Health Center, Montreal, H3A 0G4, Canada
- Division of Medical Biochemistry, McGill University Health Centre, Montréal, QC, Canada
| | - Catalin Mihalcioiu
- Department of Medical Oncology, McGill University Health Center, Montreal, Quebec, Canada
| | - Christopher E Rudd
- Départment of Medicine, Universite de Montreal, Montreal, QC, H3T 1J4, Canada.
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
- Division of Immunology-Oncology, Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, H1T 2M4, Canada.
- Division of Experimental Medicine, McGill University, Montreal, QC, H3A 0G4, Canada.
| |
Collapse
|
22
|
Sharma P, Tandel N, Kumar R, Negi S, Sharma P, Devi S, Saxena K, Chaudhary NR, Saini S, Kumar R, Chandel BS, Sijwali PS, Tyagi RK. Oleuropein activates autophagy to circumvent anti-plasmodial defense. iScience 2024; 27:109463. [PMID: 38562521 PMCID: PMC10982566 DOI: 10.1016/j.isci.2024.109463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 02/27/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Antimalarial drug resistance and unavailability of effective vaccine warrant for newer drugs and drug targets. Hence, anti-inflammatory activity of phyto-compound (oleuropein; OLP) was determined in antigen (LPS)-stimulated human THP-1 macrophages (macrophage model of inflammation; MMI). Reduction in the inflammation was controlled by the PI3K-Akt1 signaling to establish the "immune-homeostasis." Also, OLP treatment influenced the cell death/autophagy axis leading to the modulated inflammation for extended cell survival. The findings with MII prompted us to detect the antimalarial activity of OLP in the wild type (3D7), D10-expressing GFP-Atg18 parasite, and chloroquine-resistant (Dd2) parasite. OLP did not show the parasite inhibition in the routine in vitro culture of P. falciparum whereas OLP increased the antimalarial activity of artesunate. The molecular docking of autophagy-related proteins, investigations with MMI, and parasite inhibition assays indicated that the host activated the autophagy to survive OLP pressure. The challenge model of P. berghei infection showed to induce autophagy for circumventing anti-plasmodial defenses.
Collapse
Affiliation(s)
- Praveen Sharma
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh 160036, India
| | - Nikunj Tandel
- Institute of Science, Nirma University, SG highway, Ahmedabad 382481, India
| | - Rajinder Kumar
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh 160036, India
| | - Sushmita Negi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India
| | - Prakriti Sharma
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh 160036, India
| | - Sonia Devi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India
| | - Kanika Saxena
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India
- CSIR-Centre for Cellular & Molecular Biology, Hyderabad, Telangana, India
| | - Neil Roy Chaudhary
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh 160036, India
| | - Sheetal Saini
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh 160036, India
| | - Reetesh Kumar
- Faculty of Agricultural Sciences, Institute of Applied Sciences & Humanities, GLA University, Mathura 281406, India
| | - Bharat Singh Chandel
- Department of Animal Biotechnology, College of Veterinary Science and AH, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Gujarat 385 506, India
| | - Puran S. Sijwali
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India
- CSIR-Centre for Cellular & Molecular Biology, Hyderabad, Telangana, India
| | - Rajeev K. Tyagi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
23
|
Berežni S, Mimica-Dukić N, Domina G, Raimondo FM, Orčić D. Anthriscus sylvestris-Noxious Weed or Sustainable Source of Bioactive Lignans? PLANTS (BASEL, SWITZERLAND) 2024; 13:1087. [PMID: 38674496 PMCID: PMC11053937 DOI: 10.3390/plants13081087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Anthriscus sylvestris (L.) Hoffm. (Apiaceae), commonly known as wild chervil, has gained scientific interest owing to its diverse phytochemical profile and potential therapeutic applications. The plant, despite being categorized as a noxious weed, is traditionally used in treating various conditions like headaches, dressing wounds, and as a tonic, antitussive, antipyretic, analgesic, and diuretic. Its pharmacological importance stems from containing diverse bioactive lignans, especially aryltetralins and dibenzylbutyrolactones. One of the main compounds of A. sylvestris, deoxypodophyllotoxin, among its wide-ranging effects, including antitumor, antiproliferative, antiplatelet aggregation, antiviral, anti-inflammatory, and insecticidal properties, serves as a pivotal precursor to epipodophyllotoxin, crucial in the semisynthesis of cytostatic agents like etoposide and teniposide. The main starting compound for these anticancer medicines was podophyllotoxin, intensively isolated from Sinopodophyllum hexandrum, now listed as an endangered species due to overexploitation. Since new species are being investigated as potential sources, A. sylvestris emerges as a highly promising candidate owing to its abundant lignan content. This review summarizes the current knowledge on A. sylvestris, investigating its biological and morphological characteristics, and pharmacological properties. Emphasizing the biological activities and structure-activity relationship, this review underscores its therapeutic potential, thus encouraging further exploration and utilization of this valuable plant resource.
Collapse
Affiliation(s)
- Sanja Berežni
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia; (N.M.-D.); (D.O.)
| | - Neda Mimica-Dukić
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia; (N.M.-D.); (D.O.)
| | - Gianniantonio Domina
- Department of Agricultural, Food and Forest Sciences, University of Palermo, Viale delle Scienze, bldg. 4, 90128 Palermo, Italy;
| | - Francesco Maria Raimondo
- PLANTA/Center for Research, Documentation and Training, Via Serraglio Vecchio 28, 90123 Palermo, Italy;
| | - Dejan Orčić
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia; (N.M.-D.); (D.O.)
| |
Collapse
|
24
|
Hindawy RF, Manawy SM, Nafea OE, Abdelhameed AA, Hendawi FF. Moringa oleifera leaves ethanolic extract counteracts cortical neurodegeneration induced by aluminum chloride in rats. Toxicol Res (Camb) 2024; 13:tfae028. [PMID: 38455639 PMCID: PMC10917235 DOI: 10.1093/toxres/tfae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 03/09/2024] Open
Abstract
Background Aluminum, a well-recognized neurotoxin, is implicated in various neurodegenerative disorders. Moringa oleifera (M. oleifera), known as a miracle tree, is utilized as a functional food and nutritional supplement. This study investigates the potential preventive effects of M. oleifera extract on aluminum chloride (AlCl3)-induced cortical neurodegeneration in rats. Materials and methods Therefore, 24 adult male Wistar rats were randomly divided into four distinct groups: negative control, M. oleifera extract (MOE), AlCl3, and AlCl3 + MOE. Treatments were administered orally for 28 consecutive days. Cognitive performance, brain oxidative/nitrosative stress, neuroinflammation, apoptotic-cell death, and associated histopathological alterations were assessed. Results Our results showed that MOE improved spatial learning and memory, enhanced antioxidant superoxide dismutase enzyme activity, antagonized nitrosative stress, reduced inflammatory cytokines (tumor necrosis factor-alpha and interleukin-6), decreased caspase-3, increased Bcl-2, and facilitated repair of cortical and hippocampal structures. Conclusions We concluded that MOE exhibits protective effects against cortical neurodegeneration, making it a promising supplement to counteract aluminum-induced neurotoxic effects.
Collapse
Affiliation(s)
- Rabab Fawzy Hindawy
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Benha University, Al Nadi Al Ryadi, Qism Benha, Al-Qalyubia Governorate, Benha 13518, Egypt
| | - Samia M Manawy
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Al Nadi Al Ryadi, Qism Benha, Al-Qalyubia Governorate, Benha 13518, Egypt
| | - Ola Elsayed Nafea
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig Rd inside Zagazig University, Shaibet an Nakareyah, Al-Sharqia Governorate, Zagazig 44519, Egypt
| | - Abeer A Abdelhameed
- Department of Pharmacology, Faculty of Medicine, Benha University, Al Nadi Al Ryadi, Qism Benha, Al-Qalyubia Governorate, Benha 13518, Egypt
| | - Fatma Fawzi Hendawi
- Department of Pharmacology, Faculty of Medicine, Benha University, Al Nadi Al Ryadi, Qism Benha, Al-Qalyubia Governorate, Benha 13518, Egypt
| |
Collapse
|
25
|
Goldberg DJ. Induction of fat apoptosis by a combination of synchronized radiofrequency and HIFEM technology: Human histology study. J Cosmet Dermatol 2024; 23:812-817. [PMID: 38279191 DOI: 10.1111/jocd.16197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
PURPOSE With the growing demand for more effective fat reduction techniques, a combination of synchronized radiofrequency (RF) and HIFEM has been introduced. Preceding studies evidenced the ability of RF+HIFEM to maintain the fat tissue temperature at the levels necessary for adipocyte apoptosis while documenting the induced changes to the fat tissue during the several weeks after the treatment. This study aims to demonstrate the induction of apoptosis by RF+HIFEM technology in the early stages through the assessment of caspase-3 protein, one of the apoptosis-executing proteases. DESIGN In this two-arm, single-center, randomized trial, nine human subjects were enrolled and assigned into two groups, either the active group (N = 6) treated with both RF+HIFEM set at the highest tolerated levels or the sham group (N = 3) treated with 5% of the maximum RF+HIFEM power, serving as a control. All patients were scheduled to undergo one treatment visit of the abdominal area, two follow-up visits at 8 and 24 h, and one safety visit 7 days after the treatment. A punch biopsy (5 mm in diameter, approximately 10 mm in depth) was obtained from the abdominal area at the baseline and consecutive follow-up visits. Samples were fixed, and cut into 5 μm thick slices, and immunohistochemical staining was used to visualize the Caspase-3, revealing the adipocyte nuclei where apoptosis processes are in progress. FINDINGS Documented findings suggest that the temperature threshold of 43-45°C is required to initiate fat apoptosis and consequent reduction in adipocyte number was achieved during the combined treatment with RF+HIFEM. The active group showed an elevated ratio of positively stained nuclei versus all adipocyte nuclei found on the evaluated slices-referred to as the apoptotic index (AI). The AI significantly (p < 0.001) increased at both 8 h (47.01 ± 10.56%) and 24 h (43.58 ± 6.35%) posttreatment. The Sham group showed no significant change in the AI (p > 0.05). No adverse events or side effects related to the treatments were observed. SUMMARY This study supports previously published evidence on fat reduction after RF+HIFEM treatment, documenting the safe initiation of adipocyte programmed cell death posttreatment.
Collapse
Affiliation(s)
- David J Goldberg
- Skin Laser & Surgery Specialists, Division of Schwieger Dermatology Group, New York, New York, USA
- Department of Dermatology, Icahn School of Medicine at Mt. Sinai, New York, New York, USA
| |
Collapse
|
26
|
Berthenet K, Aïmontché E, El Mrini S, Brière J, Pion N, Iacono I, Brejon S, Monier K, Catez F, Ichim G, Combaret V, Mertani HC, Diaz JJ, Albaret MA. Spatial sequestration of activated-caspase 3 in aggresomes mediates resistance of neuroblastoma cell to bortezomib treatment. Sci Rep 2024; 14:3768. [PMID: 38355966 PMCID: PMC10866921 DOI: 10.1038/s41598-024-54140-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/08/2024] [Indexed: 02/16/2024] Open
Abstract
Neuroblastoma (NB) is the most common pediatric tumor and is currently treated by several types of therapies including chemotherapies, such as bortezomib treatment. However, resistance to bortezomib is frequently observed by mechanisms that remain to be deciphered. Bortezomib treatment leads to caspase activation and aggresome formation. Using models of patients-derived NB cell lines with different levels of sensitivity to bortezomib, we show that the activated form of caspase 3 accumulates within aggresomes of NB resistant cells leading to an impairment of bortezomib-induced apoptosis and increased cell survival. Our findings unveil a new mechanism of resistance to chemotherapy based on an altered subcellular distribution of the executioner caspase 3. This mechanism could explain the resistance developed in NB patients treated with bortezomib, emphasizing the potential of drugs targeting aggresomes.
Collapse
Affiliation(s)
- Kévin Berthenet
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Eliézer Aïmontché
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Sara El Mrini
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Johan Brière
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Nathalie Pion
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Isabelle Iacono
- Department of Translational Research and Innovation, Centre Léon Bérard, 69373, Lyon, France
| | - Stéphanie Brejon
- Department of Translational Research and Innovation, Centre Léon Bérard, 69373, Lyon, France
| | - Karine Monier
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Frédéric Catez
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Gabriel Ichim
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
- Institut Convergence PLAsCAN, 69373, Lyon Cedex 08, France
| | - Valérie Combaret
- Department of Translational Research and Innovation, Centre Léon Bérard, 69373, Lyon, France
| | - Hichem C Mertani
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Jean-Jacques Diaz
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
- Institut Convergence PLAsCAN, 69373, Lyon Cedex 08, France
- DevWeCan Labex Laboratory, 69373, Lyon Cedex 08, France
| | - Marie Alexandra Albaret
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France.
| |
Collapse
|
27
|
Eskandari E, Negri GL, Tan S, MacAldaz ME, Ding S, Long J, Nielsen K, Spencer SE, Morin GB, Eaves CJ. Dependence of human cell survival and proliferation on the CASP3 prodomain. Cell Death Discov 2024; 10:63. [PMID: 38321033 PMCID: PMC10847432 DOI: 10.1038/s41420-024-01826-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 02/08/2024] Open
Abstract
Mechanisms that regulate cell survival and proliferation are important for both the development and homeostasis of normal tissue, and as well as for the emergence and expansion of malignant cell populations. Caspase-3 (CASP3) has long been recognized for its proteolytic role in orchestrating cell death-initiated pathways and related processes; however, whether CASP3 has other functions in mammalian cells that do not depend on its known catalytic activity have remained unknown. To investigate this possibility, we examined the biological and molecular consequences of reducing CASP3 levels in normal and transformed human cells using lentiviral-mediated short hairpin-based knockdown experiments in combination with approaches designed to test the potential rescue capability of different components of the CASP3 protein. The results showed that a ≥50% reduction in CASP3 levels rapidly and consistently arrested cell cycle progression and survival in all cell types tested. Mass spectrometry-based proteomic analyses and more specific flow cytometric measurements strongly implicated CASP3 as playing an essential role in regulating intracellular protein aggregate clearance. Intriguingly, the rescue experiments utilizing different forms of the CASP3 protein showed its prosurvival function and effective removal of protein aggregates did not require its well-known catalytic capability, and pinpointed the N-terminal prodomain of CASP3 as the exclusive component needed in a diversity of human cell types. These findings identify a new mechanism that regulates human cell survival and proliferation and thus expands the complexity of how these processes can be controlled. The graphical abstract illustrates the critical role of CASP3 for sustained proliferation and survival of human cells through the clearance of protein aggregates.
Collapse
Affiliation(s)
- Ebrahim Eskandari
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Gian Luca Negri
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Susanna Tan
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Margarita E MacAldaz
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Shengsen Ding
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Justin Long
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Karina Nielsen
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Sandra E Spencer
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Gregg B Morin
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Connie J Eaves
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, BC, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
28
|
Kumar G, Pandey DM, Ghosh M, Dall'Acqua S, Gupta R, Tiwari NP, Siddique UM, Vishwakrama L, Guleri SK, Lal UR, Dubey S. Karanjin, A Promising Bioactive Compound Possessing Anti-cancer Activity against Experimental Model of Non-small Cell Lung Cancer Cells. Anticancer Agents Med Chem 2024; 24:317-333. [PMID: 37936467 DOI: 10.2174/0118715206255557231024095245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/07/2023] [Accepted: 09/25/2023] [Indexed: 11/09/2023]
Abstract
AIMS The aim of this study is to isolate the Millettia pinnata (Karanj) leaf extract for pure compound with anticancer properties and to study the molecular target of the isolates in non-small cell lung cancer cell lines. BACKGROUND In our earlier research Millettia pinnata leaf extract has demonstrated potential anticancer activities. Thus, in pursuit of the bioactive compounds, the most potential active extract from our previous study was purified. Furthermore, the anticancer properties of the isolated compound karanjin was studied and aimed for apoptosis and restraining growth. METHODS A novel method was developed through column chromatography for isolation and purification of the compound karanjin from leaf chloroform extract. The purified component was then characterised using FTIR, mass spectrometry, and NMR. An MTT-based cytotoxicity assay was used to analyse cell cytotoxicity, whereas fluorescence staining was used for apoptosis and reactive oxygen species inhibition quantification. Furthermore, the real-time PCR assay was used to determine the molecular mechanism of action in cells causing cytotoxicity induced by karanjin dosing. RESULTS The anticancer activity of karanjin in A549 cell line exhibited prominent activity revealing IC50 value of 4.85 μM. Conferring the predicted molecular pathway study, karanjin restrains the proliferation of cancer cells through apoptosis, which is controlled by extrinsic pathway proteins FAS/FADD/Caspases 8/3/9. Downregulation of KRAS and dependent gene expression also stopped cell proliferation. CONCLUSION Karanjin has been identified as a compound with potential effect in non-small cell lung cancer cells. Molecular mechanism for apoptosis and inhibition of reactive oxygen species induced through H2O2 were observed, concluding karanjin have medicinal and antioxidant properties.
Collapse
Affiliation(s)
- Gourav Kumar
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
- Birsa Munda Government Medical College, Shahdol, India
- Department of Pharmaceutical and Pharmacological Science, University of Padova, Italy
| | - Dev Mani Pandey
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological Science, University of Padova, Italy
| | - Rashmi Gupta
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
- Department of Pharmaceutical and Pharmacological Science, University of Padova, Italy
| | - Nishi Prakash Tiwari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Usman Mohd Siddique
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | | | | | - Uma Ranjan Lal
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
- Department of Natural Products, NIPER Ahmedabad, India
| | - Supriya Dubey
- Department of Chemistry, Kanya Gurukul Campus, Gurukul Kangri (Deemed to be University), Haridwar, India
| |
Collapse
|
29
|
Ramya Devi KT, Jaganathan MK, Ganesh MR, Dharshene K. Chitosan-encapsulated naringenin promotes ROS mediated through the activation of executioner caspase-3. Med Oncol 2023; 41:3. [PMID: 38017323 DOI: 10.1007/s12032-023-02227-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/19/2023] [Indexed: 11/30/2023]
Abstract
We previously reported that chitosan nanoparticle-encapsulated Naringenin (CS-NPs/NAR) could scavenge free radicals at lower doses and be cytotoxic to cancer cells. The current study continues to focus on the mechanism behind CS-NPs/NAR-induced breast cancer cell (MDA-MB-231) death. MDA-MB-231 cells were treated with higher concentrations (100, 200, and 200 µg) of Chitosan nanoparticles (CS-NPs), naringenin (NAR), and chitosan-encapsulated naringenin (CS-NPs/NAR). The cell viability, proliferation, and oxidative stress parameters, such as nitric oxide [NO], xanthine oxidase (XOD), and xanthine dehydrogenase (XDH) levels, were analyzed. ROS levels were determined through DCFDA analysis. MTT-based cell cytotoxicity and BrdU cell proliferation analysis depicted the cytotoxicity effects (37% and 29% for 24 and 48 h) and exhibited a reduction in the proliferation of MDA-MB-231 by CS-NPs/NAR. A significant increase in NO content, XOD, a decrease in XDH, and an increase in ROS levels were observed upon treatment with CS-NPs/NAR. Fluorescent images suggested the increase in the ROS level upon treatment with CS-NPs/NAR in cancer cells, and the results suggested that it could induce apoptosis. Further, to confirm this, the activity of caspase-3 was analyzed through western blotting, and the result suggested that the higher concentration of CS-NPs/NAR has increased the activation of procaspase3 when compared to free NAR. Hence, the current investigation concludes that high doses of CS-NPs/NAR induce and increase oxidative stress and so increased activation of procaspase3 may lead to cancer cell apoptosis and reduction in cell proliferation.
Collapse
Affiliation(s)
- K T Ramya Devi
- Faculty of Engineering and Technology, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India.
| | - M K Jaganathan
- Faculty of Engineering and Technology, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - M R Ganesh
- Department of Chemistry, College of Enginering and Technology, SRM institute of Science and Technology, Interdisciplinary Institute of Indian System of Medicine, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Karthick Dharshene
- Faculty of Engineering and Technology, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| |
Collapse
|
30
|
Dmytriv TR, Tsiumpala SA, Semchyshyn HM, Storey KB, Lushchak VI. Mitochondrial dysfunction as a possible trigger of neuroinflammation at post-traumatic stress disorder (PTSD). Front Physiol 2023; 14:1222826. [PMID: 37942228 PMCID: PMC10628526 DOI: 10.3389/fphys.2023.1222826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a neuropsychiatric disorder that occurs in approximately 15% of people as a result of some traumatic events. The main symptoms are re-experiencing and avoidance of everything related to this event and hyperarousal. The main component of the pathophysiology of PTSD is an imbalance in the functioning of the hypothalamic-pituitary-adrenal axis (HPA) and development of neuroinflammation. In parallel with this, mitochondrial dysfunction is observed, as in many other diseases. In this review, we focus on the question how mitochondria may be involved in the development of neuroinflammation and its maintaining at PTSD. First, we describe the differences in the operation of the neuro-endocrine system during stress versus PTSD. We then show changes in the activity/expression of mitochondrial proteins in PTSD and how they can affect the levels of hormones involved in PTSD development, as well as how mitochondrial damage/pathogen-associated molecule patterns (DAMPs/PAMPs) trigger development of inflammation. In addition, we examine the possibility of treating PTSD-related inflammation using mitochondria as a target.
Collapse
Affiliation(s)
- Tetiana R. Dmytriv
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Sviatoslav A. Tsiumpala
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Halyna M. Semchyshyn
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Kenneth B. Storey
- Department of Biology, Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Volodymyr I. Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
31
|
An J, Cho J. Wheat phytase potentially protects HT-29 cells from inflammatory nucleotides-induced cytotoxicity. Anim Biosci 2023; 36:1604-1611. [PMID: 37402454 PMCID: PMC10475372 DOI: 10.5713/ab.23.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/20/2023] [Accepted: 05/23/2023] [Indexed: 07/06/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate the protective effect of wheat phytase as a structural decomposer of inflammatory nucleotides, extracellular adenosine triphosphate (ATP), and uridine diphosphate (UDP) on HT-29 cells. METHODS Phosphatase activities of wheat phytase against ATP and UDP was investigated in the presence or absence of inhibitors such as L-phenylalanine and L-homoarginine using a Pi Color Lock gold phosphate detection kit. Viability of HT-29 cells exposed to intact- or dephosphorylated-nucleotides was analyzed with an EZ-CYTOX kit. Secretion levels of pro-inflammatory cytokines (IL-6 and IL-8) in HT-29 cells exposed to substrate treated with or without wheat phytase were measured with enzyme-linked immunosorbent assay kits. Activation of caspase-3 in HT-29 cells treated with intact ATP or dephosphorylated-ATP was investigated using a colorimetric assay kit. RESULTS Wheat phytase dephosphorylated both nucleotides, ATP and UDP, in a dosedependent manner. Regardless of the presence or absence of enzyme inhibitors (L-phenylalanine and L-homoarginine), wheat phytase dephosphorylated UDP. Only L-phenylalanine inhibited the dephosphorylation of ATP by wheat phytase. However, the level of inhibition was less than 10%. Wheat phytase significantly enhanced the viability of HT-29 cells against ATP- and UDP-induced cytotoxicity. Interleukin (IL)-8 released from HT-29 cells with nucleotides dephosphorylated by wheat phytase was higher than that released from HT-29 cells with intact nucleotides. Moreover, the release of IL-6 was strongly induced from HT-29 cells with UDP dephosphorylated by wheat phytase. HT-29 cells with ATP degraded by wheat phytase showed significantly (13%) lower activity of caspase-3 than HT-29 cells with intact ATP. CONCLUSION Wheat phytase can be a candidate for veterinary medicine to prevent cell death in animals. In this context, wheat phytase beyond its nutritional aspects might be a novel and promising tool for promoting growth and function of intestinal epithelial cells under luminal ATP and UDP surge in the gut.
Collapse
Affiliation(s)
- Jeongmin An
- Department of Animal Science and Technology, Konkuk University, Seoul 05029,
Korea
| | - Jaiesoon Cho
- Department of Animal Science and Technology, Konkuk University, Seoul 05029,
Korea
| |
Collapse
|
32
|
Sayres L, Flockton AR, Ji S, Rey Diaz C, Gumina DL, Su EJ. Angiogenic Function of Human Placental Endothelial Cells in Severe Fetal Growth Restriction Is Not Rescued by Individual Extracellular Matrix Proteins. Cells 2023; 12:2339. [PMID: 37830553 PMCID: PMC10572031 DOI: 10.3390/cells12192339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023] Open
Abstract
Severe fetal growth restriction (FGR) is characterized by increased placental vascular resistance resulting from aberrant angiogenesis. Interactions between endothelial cells (ECs) and the extracellular matrix (ECM) are critical to the complex process of angiogenesis. We have previously found that placental stromal abnormalities contribute to impaired angiogenesis in severe FGR. The objective of this research is to better characterize the effect of individual ECM proteins on placental angiogenic properties in the setting of severe FGR. ECs were isolated from human placentae, either control or affected by severe FGR, and subjected to a series of experiments to interrogate the role of ECM proteins on adhesion, proliferation, migration, and apoptosis. We found impaired proliferation and migration of growth-restricted ECs. Although individual substrates did not substantially impact migratory capacity, collagens I, III, and IV partially mitigated proliferative defects seen in FGR ECs. Differences in adhesion and apoptosis between control and FGR ECs were not evident. Our findings demonstrate that placental angiogenic defects that characterize severe FGR cannot be explained by a singular ECM protein, but rather, the placental stroma as a whole. Further investigation of the effects of stromal composition, architecture, stiffness, growth factor sequestration, and capacity for remodeling is essential to better understand the role of ECM in impaired angiogenesis in severe FGR.
Collapse
Affiliation(s)
- Lauren Sayres
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, CO 80045, USA
| | - Amanda R. Flockton
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, CO 80045, USA
| | - Shuhan Ji
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, CO 80045, USA
| | - Carla Rey Diaz
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, CO 80045, USA
| | - Diane L. Gumina
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, CO 80045, USA
| | - Emily J. Su
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, CO 80045, USA
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, CO 80045, USA
| |
Collapse
|
33
|
Fu Y, Deng Y, Zhang J, Chua SL, Khoo BL. Biofilms exacerbate atherogenesis through macrophage-induced inflammatory responses in a fibrous plaque microsystem model. Acta Biomater 2023; 168:333-345. [PMID: 37385520 DOI: 10.1016/j.actbio.2023.06.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
BACKGROUND Microbes have been implicated in atherosclerosis development and progression, but the impact of bacterial-based biofilms on fibrous plaque rupture remains poorly understood. RESULTS Here, we developed a comprehensive atherosclerotic model to reflect the progression of fibrous plaque under biofilm-induced inflammation (FP-I). High expressions of biofilm-specific biomarkers algD, pelA and pslB validated the presence of biofilms. Biofilm promotes the polarization of macrophages towards a pro-inflammatory (M1) phenotype, as demonstrated by an increase in M1 macrophage-specific marker CD80 expression in CD68+ macrophages. The increase in the number of intracellular lipid droplets (LDs) and foam cell percentage highlighted the potential role of biofilms on lipid synthesis or metabolic pathways in macrophage-derived foam cells. In addition, collagen I production by myofibroblasts associated with the fibrous cap was significantly reduced along with the promotion of apoptosis of myofibroblasts, indicating that biofilms affect the structural integrity of the fibrous cap and potentially undermine its strength. CONCLUSION We validated the unique role of biofilm-based inflammation in exacerbating fibrous plaque damage in the FP-I model, increasing fibrous plaque instability and risk of thrombosis. Our results lay the foundation for mechanistic studies of the role of biofilms in fibrous plaques, allowing the evaluation of preclinical combination strategies for drug therapy. STATEMENT OF SIGNIFICANCE A microsystem-based model was developed to reveal interactions in fibrous plaque during biofilm-induced inflammation (FP-I). Real-time assessment of biofilm formation and its role in fibrous plaque progression was achieved. The presence of biofilms enhanced the expression of pro-inflammatory (M1) specific marker CD80, lipid droplets, and foam cells and reduced anti-inflammatory (M2) specific marker CD206 expression. Fibrous plaque exposure to biofilm-based inflammation reduced collagen I expression and increased apoptosis marker Caspase-3 expression significantly. Overall, we demonstrate the unique role of biofilm-based inflammation in exacerbating fibrous plaque damage in the FP-I model, promoting fibrous plaque instability and enhanced thrombosis risk. Our findings lay the groundwork for mechanistic studies, facilitating the evaluation of preclinical drug combination strategies.
Collapse
Affiliation(s)
- Yatian Fu
- Department of Biomedical Engineering, City University of Hong Kong; Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE)
| | - Yanlin Deng
- Department of Biomedical Engineering, City University of Hong Kong
| | - Jing Zhang
- Department of Biomedical Engineering, City University of Hong Kong
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR China; State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR China; Shenzhen Key Laboratory of Food Biological Safety Control; Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong; Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE); City University of Hong Kong - Futian Shenzhen Research Institute.
| |
Collapse
|
34
|
Moslehi AH, Hoseinpour F, Saber A, Akhavan Taheri M, Hashemian AH. Fertility-enhancing effects of inositol & vitamin C on cisplatin induced ovarian and uterine toxicity in rats via suppressing oxidative stress and apoptosis. Food Chem Toxicol 2023; 179:113995. [PMID: 37619831 DOI: 10.1016/j.fct.2023.113995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/22/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Cisplatin can lead to infertility due to its negative impact on the uterus and ovaries. This study aimed to explore the effects of Inositol and vitamin C on cisplatin-induced infertility. Forty-eight adult female Wistar rats were divided into eight groups (N = 6) and orally treated for 21 days. The treatments were as follows: negative control (saline), positive control (saline and cisplatin injected into the abdomen on day 15), T1-T3: rats given vitamin C (150 mg/kg), Inositol (420 mg/kg), and vitamin C + Inositol, respectively, along with cisplatin injected into the abdomen on day 15, T4-T6: rats given only vitamin C, Inositol, and vitamin C + Inositol, respectively. Vitamin C and Inositol enhanced cisplatin-induced histopathological improvements in the uterus and ovaries, raising progesterone and estradiol serum levels. Furthermore, the supplements enhanced ESR1 gene expression in the uterus and ovary, reducing uterine and ovarian apoptosis caused by cisplatin through modulation of caspase 3, 8, and Bcl-2 gene levels. These substances decreased ovarian and uterine malondialdehyde levels, boosted total antioxidant capacity and superoxide dismutase, and alleviated oxidative stress. The findings reveal that vitamin C and Inositol shield against cisplatin-related infertility by reducing oxidative stress and apoptosis in the uterus and ovaries.
Collapse
Affiliation(s)
- Amir Hosein Moslehi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Fatemeh Hoseinpour
- Department of Basic Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran.
| | - Amir Saber
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Maryam Akhavan Taheri
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran; Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Hossein Hashemian
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
35
|
Chotphruethipong L, Chanvorachote P, Reudhabibadh R, Singh A, Benjakul S, Roytrakul S, Hutamekalin P. Chitooligosaccharide from Pacific White Shrimp Shell Chitosan Ameliorates Inflammation and Oxidative Stress via NF-κB, Erk1/2, Akt and Nrf2/HO-1 Pathways in LPS-Induced RAW264.7 Macrophage Cells. Foods 2023; 12:2740. [PMID: 37509832 PMCID: PMC10379839 DOI: 10.3390/foods12142740] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Chitooligosaccharide (COS), found in both insects and marine sources, has several bioactivities, such as anti-inflammation and antioxidant activities. However, the mechanism of shrimp shell COS on retardation of inflammatory and antioxidant effects is limited. Therefore, the aim of this study is to examine the mechanism of the aforementioned activities of COS in LPS-activated RAW264.7 macrophage cells. COS significantly improved cell viability in LPS-activated cells. COS at the level of 500 µg/mL could reduce the TNF-α, NO and IL-6 generations in LPS-activated cells (p < 0.05). Furthermore, COS could reduce ROS formation, NF-κB overactivation, phosphorylation of Erk1/2 and Akt and Nrf2/HO-1 in LPS-exposed cells. These results indicate that COS manifests anti-inflammatory activity and antioxidant action via NF-κB, Erk1/2, Akt and Nrf2/HO-1 signaling with an increasing relevance for inflammatory disorders.
Collapse
Affiliation(s)
- Lalita Chotphruethipong
- Department of Food Science, Faculty of Science, Burapha University, Mueang Chonburi, Chonburi 20131, Thailand
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Avtar Singh
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Thailand
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Thailand
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Pilaiwanwadee Hutamekalin
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai 90110, Thailand
| |
Collapse
|
36
|
Li G, Li J, Wang W, Feng X, Yu X, Yuan S, Zhang W, Chen J, Hu C. Synthesis, In Vitro, and In Vivo Investigations of Pterostilbene-Tethered Analogues as Anti-Breast Cancer Candidates. Int J Mol Sci 2023; 24:11468. [PMID: 37511230 PMCID: PMC10380385 DOI: 10.3390/ijms241411468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Pterostilbene has been found to be an active scaffold with anti-breast cancer (BC) action. In this study, fourteen pterostilbene-tethered analogues (2A-2N) were prepared and screened in vitro against MDA-MB-231 and MCF-7 cells. Meanwhile, their structures were characterized using 1H-NMR, 13C-NMR, and HRMS (ESI) spectroscopy techniques. Among them, analogue 2L displayed the most potent anti-proliferation effect on MDA-MB-231 (IC50 = 10.39 μM) and MCF-7 cells (IC50 = 11.73 μM). Furthermore, the meaningful structure-activity relationships suggested that the introduction of a saturated six-membered nitrogen heterocyclic ring into the side chain favored anti-BC capacity. Biological observations indicated that 2L could cause the typical morphological changes in apoptosis, namely an increase in reactive oxygen species level and a loss of mitochondrial membrane potential in BC cells. Importantly, 2L could induce mitochondrial-mediated apoptosis by regulating the expression of caspase-related proteins. Consistent with the results of our in vitro study, 2L apparently inhibited tumor growth in MDA-MB-231 xenograft mice without obvious toxicity. These findings revealed that 2L is expected to be a promising anti-BC lead compound that merits further investigations.
Collapse
Affiliation(s)
- Guoxun Li
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jian Li
- School of Pharmacy, Changzhou University, Changzhou 213164, China
- Jiangsu Key Laboratory of Advanced Catalytic Materials and Technology, Analysis and Testing Center, NERC Biomass of Changzhou University, Changzhou 213164, China
| | - Wenqian Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Xiaoqing Feng
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Xingkang Yu
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Shuo Yuan
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Wei Zhang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jialing Chen
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Caijuan Hu
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| |
Collapse
|
37
|
Magalhães HIR, Machado FA, Souza RF, Caetano MAF, Figliuolo VR, Coutinho-Silva R, Castelucci P. Study of the roles of caspase-3 and nuclear factor kappa B in myenteric neurons in a P2X7 receptor knockout mouse model of ulcerative colitis. World J Gastroenterol 2023; 29:3440-3468. [PMID: 37389242 PMCID: PMC10303518 DOI: 10.3748/wjg.v29.i22.3440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/25/2023] [Accepted: 05/12/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND The literature indicates that the enteric nervous system is affected in inflammatory bowel diseases (IBDs) and that the P2X7 receptor triggers neuronal death. However, the mechanism by which enteric neurons are lost in IBDs is unknown. AIM To study the role of the caspase-3 and nuclear factor kappa B (NF-κB) pathways in myenteric neurons in a P2X7 receptor knockout (KO) mouse model of IBDs. METHODS Forty male wild-type (WT) C57BL/6 and P2X7 receptor KO mice were euthanized 24 h or 4 d after colitis induction by 2,4,6-trinitrobenzene sulfonic acid (colitis group). Mice in the sham groups were injected with vehicle. The mice were divided into eight groups (n = 5): The WT sham 24 h and 4 d groups, the WT colitis 24 h and 4 d groups, the KO sham 24 h and 4 d groups, and the KO colitis 24 h and 4 d groups. The disease activity index (DAI) was analyzed, the distal colon was collected for immunohistochemistry analyses, and immunofluorescence was performed to identify neurons immunoreactive (ir) for calretinin, P2X7 receptor, cleaved caspase-3, total caspase-3, phospho-NF-κB, and total NF-κB. We analyzed the number of calretinin-ir and P2X7 receptor-ir neurons per ganglion, the neuronal profile area (µm²), and corrected total cell fluorescence (CTCF). RESULTS Cells double labeled for calretinin and P2X7 receptor, cleaved caspase-3, total caspase-3, phospho-NF-κB, or total NF-κB were observed in the WT colitis 24 h and 4 d groups. The number of calretinin-ir neurons per ganglion was decreased in the WT colitis 24 h and 4 d groups compared to the WT sham 24 h and 4 d groups, respectively (2.10 ± 0.13 vs 3.33 ± 0.17, P < 0.001; 2.92 ± 0.12 vs 3.70 ± 0.11, P < 0.05), but was not significantly different between the KO groups. The calretinin-ir neuronal profile area was increased in the WT colitis 24 h group compared to the WT sham 24 h group (312.60 ± 7.85 vs 278.41 ± 6.65, P < 0.05), and the nuclear profile area was decreased in the WT colitis 4 d group compared to the WT sham 4 d group (104.63 ± 2.49 vs 117.41 ± 1.14, P < 0.01). The number of P2X7 receptor-ir neurons per ganglion was decreased in the WT colitis 24 h and 4 d groups compared to the WT sham 24 h and 4 d groups, respectively (19.49 ± 0.35 vs 22.21 ± 0.18, P < 0.001; 20.35 ± 0.14 vs 22.75 ± 0.51, P < 0.001), and no P2X7 receptor-ir neurons were observed in the KO groups. Myenteric neurons showed ultrastructural changes in the WT colitis 24 h and 4 d groups and in the KO colitis 24 h group. The cleaved caspase-3 CTCF was increased in the WT colitis 24 h and 4 d groups compared to the WT sham 24 h and 4 d groups, respectively (485949 ± 14140 vs 371371 ± 16426, P < 0.001; 480381 ± 11336 vs 378365 ± 4053, P < 0.001), but was not significantly different between the KO groups. The total caspase-3 CTCF, phospho-NF-κB CTCF, and total NF-κB CTCF were not significantly different among the groups. The DAI was recovered in the KO groups. Furthermore, we demonstrated that the absence of the P2X7 receptor attenuated inflammatory infiltration, tissue damage, collagen deposition, and the decrease in the number of goblet cells in the distal colon. CONCLUSION Ulcerative colitis affects myenteric neurons in WT mice but has a weaker effect in P2X7 receptor KO mice, and neuronal death may be associated with P2X7 receptor-mediated caspase-3 activation. The P2X7 receptor can be a therapeutic target for IBDs.
Collapse
Affiliation(s)
| | | | | | | | - Vanessa Ribeiro Figliuolo
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Robson Coutinho-Silva
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | | |
Collapse
|
38
|
Vichitsakul K, Laowichuwakonnukul K, Soontornworajit B, Poomipark N, Itharat A, Rotkrua P. Anti-proliferation and induction of mitochondria-mediated apoptosis by Garcinia hanburyi resin in colorectal cancer cells. Heliyon 2023; 9:e16411. [PMID: 37292335 PMCID: PMC10245011 DOI: 10.1016/j.heliyon.2023.e16411] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction Several parts of Garcinia hanburyi are used in traditional medicine for many purposes. In this study, Garcinia hanburyi resin (GHR) was explored for possible anti-proliferative effects and the underlying mechanism on colorectal cancer (CRC) cells. Methods Gambogic acid (GA) content in GHR was analyzed by HPLC method. The cytotoxicities of GA and GHR were assessed in human CRC cell lines (SW480 and Caco-2) and normal colon cells (CCD841 CoN) using a trypan blue exclusion assay, MTS assay, and cell morphology analysis. Cell cycle and apoptosis at its half maximal inhibitory concentration (IC50) were analyzed using flow cytometry. And, the levels of intrinsic apoptosis-related proteins were measured by Western blot analysis. Results GA was the major compound as 71.26% of the GHR. The cell viability of CRC cells was decreased in a time- and dose-dependent manner after exposure to GHR. The selectivity index indicated that GHR had a high degree of selectivity against CRC cells. The same result was obtained for GA treatment. In addition, GHR markedly induced typical apoptotic morphology of CRC cells, but had no obvious effect on normal colon cells. GHR induced apoptosis with the cell cycle arrest at the G2/M phase. An increase in Bax/Bcl-2 ratio and a decrease in procaspase-3 proteins indicated that GHR promoted apoptosis by disrupting the mitochondrial outer membrane permeability and the subsequent activation of caspase-3. Conclusion GHR, which contained GA as an active compound, significantly inhibited CRC cell proliferation via the induction of intrinsic apoptosis, while having low toxicity on normal colon cells. Therefore, GHR could be proposed as a potent candidate for the treatment of CRC.
Collapse
Affiliation(s)
- Kanokkorn Vichitsakul
- Division of Biochemistry, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Khanittha Laowichuwakonnukul
- Division of Biochemistry, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Boonchoy Soontornworajit
- Department of Chemistry, Faculty of Science and Technology, Thammasat University, Pathumthani, 12120, Thailand
- Thammasat University Research Unit in Innovation of Molecular Hybrid for Biomedical Application, Pathumthani, 12120, Thailand
| | - Natwadee Poomipark
- Division of Biochemistry, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Arunporn Itharat
- Department of Applied Thai Traditional Medicine, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Pichayanoot Rotkrua
- Division of Biochemistry, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
- Thammasat University Research Unit in Innovation of Molecular Hybrid for Biomedical Application, Pathumthani, 12120, Thailand
| |
Collapse
|
39
|
Adeyemi OS, Ishii K, Kato K. L-tryptophan-titanium oxide nanoparticles showed selective anti-Toxoplasma gondii activity and improved host biocompatibility. Biomed Pharmacother 2023; 162:114597. [PMID: 36989712 DOI: 10.1016/j.biopha.2023.114597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Toxoplasma gondii, the etiological agent of toxoplasmosis, currently affects nearly one-third of the human population. Treatment options for toxoplasmosis are limited, which underscores the need for new drugs. In the present study, we screened nanoparticles (NPs) of titanium dioxide (TiO2) and molybdenum (Mo) for their potential to inhibit the growth of T. gondii in vitro. NPs of TiO2 and Mo showed non-dose-dependent anti-T. gondii activity with EC50 values of 157.6 and 253 µg/mL, respectively. Previously, we showed that amino acid modification of NPs enhances their selective anti-parasite toxicity. Therefore, to enhance the selective anti-parasitic action of TiO2, we modified the NP surface using alanine, aspartate, arginine, cysteine, glutamate, tryptophan, tyrosine, and bovine serum albumin. The bio-modified TiO2 showed anti-parasite activity with EC50 values ranging from 45.7 to 286.4 µg/mL. At effective anti-parasite concentrations, modified-TiO2 showed no appreciable host cytotoxicity. Of the eight bio-modified TiO2, tryptophan-TiO2 showed the most promising anti-T. gondii specificity and improved host biocompatibility with a selectivity index (SI) of 49.1 versus 7.5 for TiO2 (note, pyrimethamine, a standard drug for toxoplasmosis, has an SI of 2.3). Furthermore, our data indicate that redox modulation may be part of the anti-parasite action of these NPs. Indeed, augmentation with trolox and l-tryptophan reversed the growth restriction caused by the tryptophan-TiO2 NPs. Collectively, these findings suggest that the parasite toxicity was selective and not a result of general cytotoxic action. Furthermore, surface modification with amino acids such as l-tryptophan not only enhanced the anti-parasitic action of TiO2 but also improved the host biocompatibility. Overall, our findings indicate that the nutritional requirements of T. gondii represent a viable target for the development of new and effective anti-T. gondii agents.
Collapse
|
40
|
Wang S, Shan Y, Zhang S, Zhang L, Jiao Y, Xue D, Zhang L, Yi H. Lactobacillus paracasei subsp. paracasei X12 Strain Induces Apoptosis in HT-29 Cells through Activation of the Mitochondrial Pathway. Nutrients 2023; 15:2123. [PMID: 37432295 DOI: 10.3390/nu15092123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 07/12/2023] Open
Abstract
L. paracasei subsp. paracasei X12 was obtained from traditional cheese produced in northwestern China. In this study, we showed that whole peptidoglycan (WPG), extracted from L. paracasei subsp. paracasei X12, inhibited proliferation and induced apoptosis in HT-29 cells in a dose-dependent manner. In addition, WPG-induced apoptosis was associated with the loss of mitochondrial membrane potential (Ψm), the release of cytochrome c (Cyto-C) from mitochondrialto cytosolic spaces, activation of Caspase 3, and accumulation of intracellular reactive oxygen species (ROS). Finally, semi-quantitative RT-PCR showed that these events were accompanied by upregulation of proapoptotic genes (Bax or Bad) and downregulation of antiapoptotic genes (Bcl-xl). Taken together, our results demonstrated that WPG induced apoptosis in HT-29 cells through activation of the mitochondrial pathway. WPG exerted only minor toxicity upon noncancerous cells and therefore might be used as a natural agent in the treatment of cancer in future.
Collapse
Affiliation(s)
- Shumei Wang
- College of Food Engineering, Heilongjiang Province Key Laboratory of Cold Region Wetland Ecology and Environment Research, Harbin University, Harbin 150086, China
| | - Yi Shan
- College of Food Engineering, Heilongjiang Province Key Laboratory of Cold Region Wetland Ecology and Environment Research, Harbin University, Harbin 150086, China
| | - Shuang Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China
| | - Yuehua Jiao
- Center of Drug Safety Evaluation, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Dijia Xue
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lili Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China
| |
Collapse
|
41
|
Abd-Allah ER, Fouad NY, Ghareeb AEWE, Eldebss TMA. Chloroacetonitrile reduces rat prenatal bone length and induces oxidative stress, apoptosis, and DNA damage in rat fetal liver. Birth Defects Res 2023; 115:614-632. [PMID: 36751045 DOI: 10.1002/bdr2.2155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 01/15/2023] [Accepted: 01/24/2023] [Indexed: 02/09/2023]
Abstract
One of the disinfection byproducts of chlorinating drinking water is chloroacetonitrile (CAN). Thirty-six female rats were used and distributed equally into four groups. The low dose treated group received CAN at a dose of 5.5 mg/kg body weight/day (1/40 LD50 ) orally from the 6th to 12th day of gestation. The high dose treated group received 11 mg/kg body weight/day (1/20 LD50 ) of CAN orally for the same period, the vehicle control group received 1 mL of corn oil, and the water control group received 1 mL of distilled water orally for the same period. High dose exposure to CAN significantly reduced gravid uterine weight, fetal body weights, and length, and caused obvious skeletal deformities, weak mineralization. Fetal tibial growth plates displayed histopathologic changes. Induced oxidative stress and redox imbalance in fetal liver tissues was evidenced by significantly decreased in catalase and superoxide dismutase activity, and elevated malondialdehyde levels. Histopathological, glycogen content changes, and DNA damage were observed in the fetal liver of high dose treated group. Additionally, administration of high dose of CAN induced apoptosis, evidenced by increased caspase-3 concentration in fetal liver. Thus, extensive exposure to CAN induces poor pregnancy outcomes. CAN levels in water should be monitored regularly.
Collapse
Affiliation(s)
- Entsar R Abd-Allah
- Department of Zoology, Faculty of Science, Al-Azhar University, Nasr City, Egypt
| | - Nourhan Y Fouad
- Department of Biotechnology, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Taha M A Eldebss
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
42
|
Zarnitz L, Doorschodt BM, Ernst L, Hosseinnejad A, Edgworth E, Fechter T, Theißen A, Djudjaj S, Boor P, Rossaint R, Tolba RH, Bleilevens C. Taurine as Antioxidant in a Novel Cell- and Oxygen Carrier-Free Perfusate for Normothermic Machine Perfusion of Porcine Kidneys. Antioxidants (Basel) 2023; 12:antiox12030768. [PMID: 36979015 PMCID: PMC10045130 DOI: 10.3390/antiox12030768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Donor organ-shortage has resulted in the increased use of marginal grafts; however, normothermic machine perfusion (NMP) holds the potential for organ viability assessment and restoration of marginal grafts prior to transplantation. Additionally, cell-, oxygen carrier-free and antioxidants-supplemented solutions could potentially prevent adverse effects (transfusion reactions, inflammation, hemolysis), associated with the use of autologous packed red blood cell (pRBC)-based perfusates. This study compared 6 h NMP of porcine kidneys, using an established pRBC-based perfusate (pRBC, n = 7), with the novel cell- and oxygen carrier-free organ preservation solution Ecosol, containing taurine (Ecosol, n = 7). Despite the enhanced tissue edema and tubular injury in the Ecosol group, related to a suboptimal molecular mass of polyethylene glycol as colloid present in the solution, functional parameters (renal blood flow, intrarenal resistance, urinary flow, pH) and oxygenation (arterial pO2, absence of hypoxia-inducible factor 1-alpha) were similar to the pRBC group. Furthermore, taurine significantly improved the antioxidant capacity in the Ecosol group, reflected in decreased lactate dehydrogenase, urine protein and tubular vacuolization compared to pRBC. This study demonstrates the feasibility of 6 h NMP using a taurine containing, cell- and oxygen carrier-free perfusate, achieving a comparable organ quality to pRBC perfused porcine kidneys.
Collapse
Affiliation(s)
- Laura Zarnitz
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Benedict M Doorschodt
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Lisa Ernst
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Aisa Hosseinnejad
- DWI-Leibniz-Institute for Interactive Materials e.V., 52056 Aachen, Germany
| | - Eileen Edgworth
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Tamara Fechter
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Alexander Theißen
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Sonja Djudjaj
- Institute of Pathology & Division of Nephrology, Medical Faculty, RWTH Aachen, 52074 Aachen, Germany
| | - Peter Boor
- Institute of Pathology & Division of Nephrology, Medical Faculty, RWTH Aachen, 52074 Aachen, Germany
| | - Rolf Rossaint
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - René H Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Christian Bleilevens
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| |
Collapse
|
43
|
Adeyemi OS, Ishii K, Kato K. The In Vitro Anti-Parasitic Activities of Emodin toward Toxoplasma gondii. Pharmaceuticals (Basel) 2023; 16:ph16030447. [PMID: 36986545 PMCID: PMC10053859 DOI: 10.3390/ph16030447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Currently, toxoplasmosis affects nearly one-third of the world’s population, but the available treatments have several limitations. This factor underscores the search for better therapy for toxoplasmosis. Therefore, in the current investigation, we investigated the potential of emodin as a new anti-Toxoplasma gondii while exploring its anti-parasitic mechanism of action. We explored the mechanisms of action of emodin in the presence and absence of an in vitro model of experimental toxoplasmosis. Emodin showed strong anti-T. gondii action with an EC50 value of 0.03 µg/mL; at this same effective anti-parasite concentration, emodin showed no appreciable host cytotoxicity. Likewise, emodin showed a promising anti-T. gondii specificity with a selectivity index (SI) of 276. Pyrimethamine, a standard drug for toxoplasmosis, had an SI of 2.3. The results collectively imply that parasite damage was selective rather than as a result of a broad cytotoxic effect. Furthermore, our data confirm that emodin-induced parasite growth suppression stems from parasite targets and not host targets, and indicate that the anti-parasite action of emodin precludes oxidative stress and ROS production. Emodin likely mediates parasite growth suppression through means other than oxidative stress, ROS production, or mitochondrial toxicity. Collectively, our findings support the potential of emodin as a promising and novel anti-parasitic agent that warrants further investigation.
Collapse
Affiliation(s)
- Oluyomi Stephen Adeyemi
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, 232-3 Yomogida, Naruko-onsen, Osaki 989-6711, Miyagi, Japan
- Medicinal Biochemistry and Toxicology Laboratory, Department of Biochemistry, Landmark University, Omu-Aran 251101, Kwara State, Nigeria
| | - Kosei Ishii
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, 232-3 Yomogida, Naruko-onsen, Osaki 989-6711, Miyagi, Japan
| | - Kentaro Kato
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, 232-3 Yomogida, Naruko-onsen, Osaki 989-6711, Miyagi, Japan
- Correspondence: ; Tel./Fax: +81-229-84-7391
| |
Collapse
|
44
|
Ahmed RA, Alam MF, Alshahrani S, Jali AM, Qahl AM, Khalid M, Muzafar HMA, Alhamami HN, Anwer T. Capsaicin Ameliorates the Cyclophosphamide-Induced Cardiotoxicity by Inhibiting Free Radicals Generation, Inflammatory Cytokines, and Apoptotic Pathway in Rats. Life (Basel) 2023; 13:life13030786. [PMID: 36983940 PMCID: PMC10056591 DOI: 10.3390/life13030786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/02/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Cyclophosphamide is an antineoplastic agent that has a broad range of therapeutic applications; however, it has numerous side effects, including cardiotoxicity. Furthermore, chili peppers contain a substance called capsaicin, having antioxidant and anti-inflammatory effects. Thus, this research paper focuses on the potential mechanism of capsaicin’s cardioprotective activity against cyclophosphamide-induced cardiotoxicity by measuring the expression of oxidative and inflammatory marker such as interleukins and caspases. The following groups of rats were randomly assigned: only vehicle given for 6 days (control group); cyclophosphamide 200 mg/kg intraperitoneal on 4th day only (positive control group); capsaicin 10 mg/kg orally given for 6 days followed by cyclophosphamide 200 mg/kg on 4th day of treatment; capsaicin 20 mg/kg orally for six days followed by cyclophosphamide 200 mg/kg on 4th day of treatment; and maximum amount of capsaicin alone (20 mg/kg) orally for six days. Using ELISA kits, it was found that the cyclophosphamide administration significantly increased the levels of lactate dehydrogenase, troponin-I (cardiac cell damage marker), lipid peroxidation, triglyceride, interleukin-6, tumor necrosis factor-alpha, and caspase 3. However, it markedly reduced the antioxidant enzymes catalase and glutathione levels. Both doses of capsaicin could reverse cardiac cell damage markers, as shown by a significant decline in (lactate dehydrogenase and troponin-I). In addition, capsaicin significantly reduced the cytokine levels (interleukin-6 and tumor necrosis factor-alpha), caspase 3, lipid peroxidation, and triglycerides. However, capsaicin treatment significantly raised the antioxidant content of enzymes such as glutathione and catalase. The capsaicin-treated group restored the oxidative parameter’s imbalance and generated considerable protection against cardiomyocyte harm from cyclophosphamide in male Wistar rats. These protective effects might be beneficial against the negative impacts of cyclophosphamide when used to treat cancer and immune-mediated diseases.
Collapse
Affiliation(s)
- Rayan A. Ahmed
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
- Correspondence: (R.A.A.); (M.F.A.)
| | - Mohammad Firoz Alam
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
- Correspondence: (R.A.A.); (M.F.A.)
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Abdulmajeed M. Jali
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Abdullah M. Qahl
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 16278, Saudi Arabia
| | - Hisham M. A. Muzafar
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Hussain N. Alhamami
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Tarique Anwer
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
45
|
Han HH, Wang HM, Jangili P, Li M, Wu L, Zang Y, Sedgwick AC, Li J, He XP, James TD, Kim JS. The design of small-molecule prodrugs and activatable phototherapeutics for cancer therapy. Chem Soc Rev 2023; 52:879-920. [PMID: 36637396 DOI: 10.1039/d2cs00673a] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Cancer remains as one of the most significant health problems, with approximately 19 million people diagnosed worldwide each year. Chemotherapy is a routinely used method to treat cancer patients. However, current treatment options lack the appropriate selectivity for cancer cells, are prone to resistance mechanisms, and are plagued with dose-limiting toxicities. As such, researchers have devoted their attention to developing prodrug-based strategies that have the potential to overcome these limitations. This tutorial review highlights recently developed prodrug strategies for cancer therapy. Prodrug examples that provide an integrated diagnostic (fluorescent, photoacoustic, and magnetic resonance imaging) response, which are referred to as theranostics, are also discussed. Owing to the non-invasive nature of light (and X-rays), we have discussed external excitation prodrug strategies as well as examples of activatable photosensitizers that enhance the precision of photodynamic therapy/photothermal therapy. Activatable photosensitizers/photothermal agents can be seen as analogous to prodrugs, with their phototherapeutic properties at a specific wavelength activated in the presence of disease-related biomarkers. We discuss each design strategy and illustrate the importance of targeting biomarkers specific to the tumour microenvironment and biomarkers that are known to be overexpressed within cancer cells. Moreover, we discuss the advantages of each approach and highlight their inherent limitations. We hope in doing so, the reader will appreciate the current challenges and available opportunities in the field and inspire subsequent generations to pursue this crucial area of cancer research.
Collapse
Affiliation(s)
- Hai-Hao Han
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, P. R. China. .,State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China.,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, P. R. China
| | - Han-Min Wang
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Paramesh Jangili
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| | - Mingle Li
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| | - Luling Wu
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK.
| | - Yi Zang
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,Lingang laboratory, Shanghai 201203, China
| | - Adam C Sedgwick
- Chemistry Research Laboratory, University of Oxford, Mansfield Road, OX1 3TA, UK.
| | - Jia Li
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China.,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, P. R. China
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, P. R. China. .,The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China.,National Center for Liver Cancer, Shanghai 200438, China
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK. .,School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
46
|
Choi J, Yoo MJ, Park SY, Seol JW. Antitumor Effects of Esculetin, a Natural Coumarin Derivative, against Canine Mammary Gland Tumor Cells by Inducing Cell Cycle Arrest and Apoptosis. Vet Sci 2023; 10:vetsci10020084. [PMID: 36851388 PMCID: PMC9961495 DOI: 10.3390/vetsci10020084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Mammary gland tumors are the most common neoplasms in female dogs, of which 50% are malignant. Esculetin, a coumarin derivative, reportedly induces death in different types of cancer cells. In this study, we explore the anticancer effects of esculetin against CMT-U27 and CF41.mg canine mammary gland tumor cells. Esculetin significantly inhibited the viability and migration of both CMT-U27 and CF41.mg cells in a dose- and time-dependent manner. Flow cytometric analysis and terminal deoxynucleotidyl transferase dUTP nick-end labeling assay revealed increased numbers of annexin-V-positive cells and DNA fragmentation. Furthermore, a cell cycle analysis demonstrated that esculetin blocked the cell progression at the G0/G1 phase and the S phase in CMT-U27 and CF41.mg cells. These results were supported by a Western blot analysis, which revealed upregulated protein expression of cleaved caspase-3, a marker of apoptosis, and downregulated cyclin-dependent kinase 4 and cyclin D1 protein, the cell cycle regulators. In conclusion, this novel study proves that esculetin exerts in vitro antitumor effects by inducing apoptosis and cell cycle arrest in canine mammary gland tumors.
Collapse
Affiliation(s)
- Jawun Choi
- Correspondence: (J.C.); (J.-W.S.); Tel.: +82-63-850-0964 (J.-W.S.)
| | | | | | - Jae-Won Seol
- Correspondence: (J.C.); (J.-W.S.); Tel.: +82-63-850-0964 (J.-W.S.)
| |
Collapse
|
47
|
Ashour WM, Zamzam MSA, Sayed Ali HEEDE, Ebrahim RH. Effect of fetuin-A on adenine-induced chronic kidney disease model in male rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:511-516. [PMID: 37051102 PMCID: PMC10083833 DOI: 10.22038/ijbms.2023.66346.14584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 02/07/2023] [Indexed: 04/14/2023]
Abstract
Objectives This study aimed to investigate the possible effects of fetuin-A on an adenine-induced chronic kidney disease (CKD) model in male rats. Materials and Methods Rats were divided into three groups: group A included rats fed a normal diet; group B included rats fed a normal diet with 220 mg/kg adenine daily for 21 days; group C included rats fed a normal diet with 220 mg/kg adenine daily for 21 days and intraperitoneally administered with 5 mg\kg fetuin-A every other day for 2 weeks. Serum samples were assayed for serum creatinine, urea, sodium, potassium, calcium, phosphorus, tumor necrosis factor (TNF), interleukin-6 (IL-6), and estimated glomerular filtration rate (eGFR), and immunohistochemical staining was performed. Results Group B showed a significant increase in serum creatinine, urea, phosphorus, potassium, TNF, and IL-6 and a significant decrease in serum sodium, calcium, and eGFR compared with group A. Regarding immunohistochemistry, group B showed increased apoptosis. In group C, fetuin-A reduced the urea, creatinine, and phosphorus levels, and in group C, fetuin-A decreased inflammation and apoptosis by reduction of caspase-3 staining. Conclusion Fetuin-A improved kidney function in CKD due to its anti-inflammatory and anti-fibrotic role.
Collapse
Affiliation(s)
- Wesam M.R Ashour
- Physiology Department, Faculty of Medicine, Zagazig university, Sharqia government, Egypt
| | | | | | - Reham Hassan Ebrahim
- Physiology Department, Faculty of Medicine, Zagazig university, Sharqia government, Egypt
- Corresponding author: Reham Hassan Ebrahim. Physiology Department, Faculty of Medicine, Zagazig university, Sharqia government, Egypt.
| |
Collapse
|
48
|
Wang F, Yan X, Hua Y, Song J, Liu D, Yang C, Peng F, Kang F, Hui Y. PI3K/AKT/mTOR pathway and its related molecules participate in PROK1 silence-induced anti-tumor effects on pancreatic cancer. Open Life Sci 2023; 18:20220538. [PMID: 37070074 PMCID: PMC10105552 DOI: 10.1515/biol-2022-0538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/28/2022] [Accepted: 11/21/2022] [Indexed: 04/19/2023] Open
Abstract
The PI3K/AKT/mTOR (phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin) pathway can be initiated by PROK1 (prokineticin 1), but its effect and mechanism of action in pancreatic carcinoma (PC) are not fully understood. In this study, we elucidated the roles of PROK1 and its related molecules in PC in vivo. PANC-1 cells with PROK1 knockdown were injected into BALB/c nude mice. The growth and weight of the tumor were monitored and measured, which was followed by TUNEL (terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling), immunohistochemical staining, and hematoxylin and eosin staining. The key proteins related to proliferation, apoptosis, and the PI3K/AKT/mTOR pathway were determined by Western blotting. We also used public databases to identify the molecules related to PROK1. The reduction of PROK1 inhibited angiopoiesis and promoted apoptosis in vivo. PCNA-1, cyclin D1, and Bcl-2 decreased considerably, while Bax and cleaved caspase-3 increased significantly after PROK1 inhibition. The PI3K/AKT/mTOR signal inhibition was also closely associated with PROK1 knockdown. The possible related molecules of PROK1, such as von Willebrand factor, were screened and considered to be involved in the aberrant activation of PI3K/AKT. In conclusion, PROK1 knockdown significantly prevented tumor growth and promoted apoptosis of human PC cells in vivo, where the PI3K/AKT/mTOR pathway was probably inhibited. Therefore, PROK1, along with its related molecules, might be important targets for PC therapy.
Collapse
Affiliation(s)
- Feng Wang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Xiaogang Yan
- Department of Surgical Oncology, The First People’s Hospital of Yinchuan, Yinchuan750001, China
| | - Yongqiang Hua
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, Shanghai200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Jianjun Song
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Di Liu
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Chun Yang
- Department of Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan750001, China
| | - Fei Peng
- Department of Hepatobiliary Pancreatic Surgery, Edong Healthcare Huangshi Central Hospital, Huangshi435002, Hubei, China
| | - Fuping Kang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Yongfeng Hui
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| |
Collapse
|
49
|
Wei S, Feng M, Zhang S. Molecular Characteristics of Cell Pyroptosis and Its Inhibitors: A Review of Activation, Regulation, and Inhibitors. Int J Mol Sci 2022; 23:ijms232416115. [PMID: 36555757 PMCID: PMC9783510 DOI: 10.3390/ijms232416115] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/02/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Pyroptosis is an active and ordered form of programmed cell death. The signaling pathways of pyroptosis are mainly divided into canonical pathways mediated by caspase-1 and noncanonical pathways mediated by caspase-11. Cell pyroptosis is characterized by the activation of inflammatory caspases (mainly caspase-1, 4, 5, 11) and cleavage of various members of the Gasdermin family to form membrane perforation components, leading to cell membrane rupture, inflammatory mediators release, and cell death. Moderate pyroptosis is an innate immune response that fights against infection and plays an important role in the occurrence and development of the normal function of the immune system. However, excessive pyroptosis occurs and leads to immune disorders in many pathological conditions. Based on canonical pathways, research on pyroptosis regulation has demonstrated several pyroptotic inhibitors, including small-molecule drugs, natural products, and formulations of traditional Chinese medicines. In this paper, we review the characteristics and molecular mechanisms of pyroptosis, summarize inhibitors of pyroptosis, and propound that herbal medicines should be a focus on the research and development for pyroptosis blockers.
Collapse
Affiliation(s)
| | | | - Shidong Zhang
- Correspondence: ; Tel.: +86-931-211-5256; Fax: +86-931-211-5191
| |
Collapse
|
50
|
Cicek B, Genc S, Yeni Y, Kuzucu M, Cetin A, Yildirim S, Bolat I, Kantarci M, Hacimuftuoglu A, Lazopoulos G, Tsatsakis A, Tsarouhas K, Taghizadehghalehjoughi A. Artichoke (Cynara Scolymus) Methanolic Leaf Extract Alleviates Diethylnitrosamine-Induced Toxicity in BALB/c Mouse Brain: Involvement of Oxidative Stress and Apoptotically Related Klotho/PPARγ Signaling. J Pers Med 2022; 12:2012. [PMID: 36556233 PMCID: PMC9781370 DOI: 10.3390/jpm12122012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/07/2022] Open
Abstract
(1) Background: Various epidemiological studies suggest that oxidative stress and disrupted neuronal function are mechanistically linked to neurodegenerative diseases (NDs), including Parkinson's disease (PD) and Alzheimer's disease (AD). DNA damage, oxidative stress, lipid peroxidation, and eventually, cell death such as NDs can be induced by nitrosamine-related compounds, leading to neurodegeneration. A limited number of studies have reported that exposure to diethylnitrosamine (DEN), which is commonly found in processed/preserved foods, causes biochemical abnormalities in the brain. Artichoke leaves have been used in traditional medicine as a beneficial source of bioactive components such as hydroxycinnamic acids, cynarine, chlorogenic acid, and flavonoids (luteolin and apigenin). The aim of this study is to investigate the favorable effects of exogenous artichoke (Cynara scolymus) methanolic leaf extract supplementation in ameliorating DEN-induced deleterious effects in BALB/c mouse brains. (2) Methods: This study was designed to evaluate DEN (toxicity induction by 100 mg/kg) and artichoke (protective effects of 0.8 and 1.6 g/kg treatment) for 14 days. All groups underwent a locomotor activity test to evaluate motor activity. In brain tissue, oxidative stress indicators (TAC, TOS, and MDA), Klotho and PPARγ levels, and apoptotic markers (Bax, Bcl-2, and caspase-3) were measured. Brain slices were also examined histopathologically. (3) Results: Artichoke effectively ameliorated DEN-induced toxicity with increasing artichoke dose. Impaired motor function and elevated oxidative stress markers (decreasing MDA and TOS levels and increasing TAC level) induced by DEN intoxication were markedly restored by high-dose artichoke treatment. Artichoke significantly improved the levels of Klotho and PPARγ, which are neuroprotective factors, in mouse brain tissue exposed to DEN. In addition, caspase-3 and Bax levels were reduced, whereas the Bcl-2 level was elevated with artichoke treatment. Furthermore, recovery was confirmed by histopathological analysis. (4) Conclusions: Artichoke exerted neuroprotective effects against DEN-induced brain toxicity by mitigating oxidant parameters and exerting antioxidant and antiapoptotic effects. Further research is needed to fully identify the favorable impact of artichoke supplementation on all aspects of DEN brain intoxication.
Collapse
Affiliation(s)
- Betul Cicek
- Department of Physiology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey
| | - Sidika Genc
- Department of Medical Pharmacology, Faculty of Medicine, Bilecik Seyh Edebali University, Bilecik 11230, Turkey
| | - Yesim Yeni
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum 25240, Turkey
| | - Mehmet Kuzucu
- Department of Biology, Faculty of Arts and Sciences, Erzincan Binali Yildirim University, Erzincan 24100, Turkey
| | - Ahmet Cetin
- Department of Biology, Graduate School of Natural and Applied Sciences, Erzincan Binali Yildirim University, Erzincan 24100, Turkey
| | - Serkan Yildirim
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum 25240, Turkey
| | - Ismail Bolat
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum 25240, Turkey
| | - Mecit Kantarci
- Department of Radiology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey
- Department of Radiology, Faculty of Medicine, Ataturk University, Erzurum 25240, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum 25240, Turkey
| | - Georgios Lazopoulos
- Department of Cardiac Surgery, University General Hospital of Heraklion, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | | | - Ali Taghizadehghalehjoughi
- Department of Medical Pharmacology, Faculty of Medicine, Bilecik Seyh Edebali University, Bilecik 11230, Turkey
| |
Collapse
|