1
|
Qin S, Zhu Y, Tian G, Jensen MB, Zhang K, Ding X, Bai S, Wang J, Xuan Y, Zeng Q. Dietary resistant starch protects against post-antibiotic intestinal damage by restoring microbial homeostasis and preserving intestinal barrier function in meat duck. Poult Sci 2025; 104:105213. [PMID: 40294558 DOI: 10.1016/j.psj.2025.105213] [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: 02/12/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025] Open
Abstract
Resistant starch (RS) is recognized as a nutritional strategy that supports gut and overall host health by modulating gut microbiota. To directly assess the effects of RS on gut microbiota and its role in improving intestinal barrier function in meat ducks, this study first established an antibiotic-induced microbial dysbiosis model, which was characterized by reduced gut microbial diversity, intestinal dysfunction, and an inflammatory outburst following antibiotic exposure. Whereafter, in addition to the control group, ducks treated with antibiotics for 7 consecutive days were further allocated to two groups and fed the basal diet and RS diet that derived from 12 % raw potato starch until 21 d. The results demonstrated that dietary RS supplementation reversed the antibiotic-induced reduction in microbial diversity and restored the Firmicutes-to-Bacteroidetes ratio. Additionally, RS inclusion enriched beneficial bacterial genera, including Coprobacter, Odoribacter, and Faecalibacterium (LDA score > 3). Post-antibiotic intervention led to a reduction in villus density and muscular thickness, accompanied by a significant downregulation (P < 0.05) of zonula occludens-1 and mucin-2 expression, along with increased serum pro-inflammatory cytokine levels (P < 0.05). Notably, dietary RS supplementation significantly enhanced (P < 0.05) the expression of glucagon-like peptide receptor and the anti-apoptotic factor Bcl-2, while suppressing caspase transcription. This resulted in increased villus height and muscular thickness in the ileum (P < 0.05). Furthermore, RS intervention remarkably reduced (P < 0.05) pro-inflammatory cytokine levels, particularly interleukin-1β and tumor necrosis factor-α, in both the ileum and serum. These effects were likely linked to alterations in cecal microbiota, including increased abundances of Barnesiella, Ruminiclostridium 9, Megamonas, Faecalitalea, Adlercreutzia, Coprobacter and Collinsella. In conclusion, dietary RS supplementation mitigated antibiotic-induced cecal microbial dysbiosis and restored intestinal structure by promoting enterocyte proliferation and reducing apoptosis. Consequently, RS supplementation helped alleviate systemic inflammation in meat ducks following antibiotic treatment.
Collapse
Affiliation(s)
- Simeng Qin
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, China; College of Animal Science and Technology, Southwest University, Rongchang, Chongqing, China
| | - Yifeng Zhu
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Gang Tian
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | | | - Keying Zhang
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Xuemei Ding
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Shiping Bai
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Jianping Wang
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Yue Xuan
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Qiufeng Zeng
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, China.
| |
Collapse
|
2
|
Cai JL, Zhang Y, Gao H, Wang Q, Huang W, Cai YJ, Jia WX, Wang JJ, Chen X, Sun HY. Molecular characterization, expression pattern and the function of TRAF2 from blood parrot Amphilophus citrinellus ×Vieja melanura response to LPS stimulation. FISH & SHELLFISH IMMUNOLOGY 2025; 163:110362. [PMID: 40280260 DOI: 10.1016/j.fsi.2025.110362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
Tumor necrosis factor receptor-associated factor (TRAF) family is a critical signal transduction protein, and plays important roles in cell growth, apoptosis, and immune response, etc. In this study, molecular characteristics, expression patterns, and the role of TRAF2 in blood parrot Vieja synspila ♀ × Amphilophus citrinellus ♂, an important ornamental fish, were explored response to lipopolysaccharide (LPS) challenge. The full length of blood parrot TRAF2 was 2725 bp, with an open reading frame (ORF) of 1551 bp encoding 516 amino acids, and a molecular weight of 58.58 kDa. Blood parrot TRAF2 contained four conserved domains: RING, TRAF-type zinc finger, TRAF_BIRC3_bd, and MATH (Meprin and TRAF-C homology). Analysis of phylogenetic relationships showed that TRAF2 were conserved in different species, indicating that its role might be similar. Blood parrot TRAF2 mRNA could be detected in all of the tissues examined, and was distributed in both the cytoplasm and nucleus. The expression of blood parrot TRAF2 was up-regulated during LPS challenge. Overexpression of TRAF2 could significantly inhibit the activities of nuclear factor κB (NF-κB) and activated protein 1 (AP-1), and reduce the ratio of Bax/Bcl-2. This study indicated that the TRAF2 might play important roles in organisms during pathogen infection.
Collapse
Affiliation(s)
- Jie-Li Cai
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; School of Life Sciences, South China Normal University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yue Zhang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Hui Gao
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Qi Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wei Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yi-Jie Cai
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wei-Xin Jia
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jun-Jie Wang
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong, China.
| | - Xiao Chen
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.
| | - Hong-Yan Sun
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Vogler M, Braun Y, Smith VM, Westhoff MA, Pereira RS, Pieper NM, Anders M, Callens M, Vervliet T, Abbas M, Macip S, Schmid R, Bultynck G, Dyer MJ. The BCL2 family: from apoptosis mechanisms to new advances in targeted therapy. Signal Transduct Target Ther 2025; 10:91. [PMID: 40113751 PMCID: PMC11926181 DOI: 10.1038/s41392-025-02176-0] [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: 11/07/2024] [Revised: 12/21/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
The B cell lymphoma 2 (BCL2) protein family critically controls apoptosis by regulating the release of cytochrome c from mitochondria. In this cutting-edge review, we summarize the basic biology regulating the BCL2 family including canonical and non-canonical functions, and highlight milestones from basic research to clinical applications in cancer and other pathophysiological conditions. We review laboratory and clinical development of BH3-mimetics as well as more recent approaches including proteolysis targeting chimeras (PROTACs), antibody-drug conjugates (ADCs) and tools targeting the BH4 domain of BCL2. The first BCL2-selective BH3-mimetic, venetoclax, showed remarkable efficacy with manageable toxicities and has transformed the treatment of several hematologic malignancies. Following its success, several chemically similar BCL2 inhibitors such as sonrotoclax and lisaftoclax are currently under clinical evaluation, alone and in combination. Genetic analysis highlights the importance of BCL-XL and MCL1 across different cancer types and the possible utility of BH3-mimetics targeting these proteins. However, the development of BH3-mimetics targeting BCL-XL or MCL1 has been more challenging, with on-target toxicities including thrombocytopenia for BCL-XL and cardiac toxicities for MCL1 inhibitors precluding clinical development. Tumor-specific BCL-XL or MCL1 inhibition may be achieved by novel targeting approaches using PROTACs or selective drug delivery strategies and would be transformational in many subtypes of malignancy. Taken together, we envision that the targeting of BCL2 proteins, while already a success story of translational research, may in the foreseeable future have broader clinical applicability and improve the treatment of multiple diseases.
Collapse
Affiliation(s)
- Meike Vogler
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) partner site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany.
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany.
| | - Yannick Braun
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Victoria M Smith
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Raquel S Pereira
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Nadja M Pieper
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Marius Anders
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Manon Callens
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Tim Vervliet
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Maha Abbas
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Salvador Macip
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Josep Carreras Leukaemia Research Institute, Badalona, Spain
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Ralf Schmid
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Institute for Structural and Chemical Biology, University of Leicester, Leicester, UK
| | - Geert Bultynck
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Martin Js Dyer
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
4
|
Poole K, Iyer KS, Schmidtke DW, Petroll WM, Varner VD. Corneal Keratocytes, Fibroblasts, and Myofibroblasts Exhibit Distinct Transcriptional Profiles In Vitro. Invest Ophthalmol Vis Sci 2025; 66:28. [PMID: 40072446 PMCID: PMC11918030 DOI: 10.1167/iovs.66.3.28] [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] [Indexed: 03/14/2025] Open
Abstract
Purpose After stromal injury to the cornea, the release of growth factors and pro-inflammatory cytokines promotes the activation of quiescent keratocytes into a migratory fibroblast and/or fibrotic myofibroblast phenotype. Persistence of the myofibroblast phenotype can lead to corneal fibrosis and scarring, which are leading causes of blindness worldwide. This study aims to establish comprehensive transcriptional profiles for cultured corneal keratocytes, fibroblasts, and myofibroblasts to gain insights into the mechanisms through which these phenotypic changes occur. Methods Primary rabbit corneal keratocytes were cultured in either defined serum-free (SF) media, fetal bovine serum (FBS) containing media, or SF media supplemented with TGF-β1 to induce keratocyte, fibroblast, or myofibroblast phenotypes, respectively. Bulk RNA sequencing followed by bioinformatic analyses was performed to identify significant differentially expressed genes (DEGs) and enriched biological pathways for each phenotype. Results Genes commonly associated with keratocytes, fibroblasts, or myofibroblasts showed high relative expression in SF, FBS, or TGF-β1 culture conditions, respectively. Differential expression and functional analyses revealed novel DEGs for each cell type, as well as enriched pathways indicative of differences in proliferation, apoptosis, extracellular matrix (ECM) synthesis, cell-ECM interactions, cytokine signaling, and cell mechanics. Conclusions Overall, these data demonstrate distinct transcriptional differences among cultured corneal keratocytes, fibroblasts, and myofibroblasts. We have identified genes and signaling pathways that may play important roles in keratocyte differentiation, including many related to mechanotransduction and ECM biology. Our findings have revealed novel molecular markers for each cell type, as well as possible targets for modulating cell behavior and promoting physiological corneal wound healing.
Collapse
Affiliation(s)
- Kara Poole
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Krithika S Iyer
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas, United States
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, Texas, United States
| | - David W Schmidtke
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas, United States
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, Texas, United States
| | - W Matthew Petroll
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, Texas, United States
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Victor D Varner
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas, United States
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
5
|
Earnhardt-San AL, Baker EC, Cilkiz KZ, Cardoso RC, Ghaffari N, Long CR, Riggs PK, Randel RD, Riley DG, Welsh TH. Evaluation of Prenatal Transportation Stress on DNA Methylation (DNAm) and Gene Expression in the Hypothalamic-Pituitary-Adrenal (HPA) Axis Tissues of Mature Brahman Cows. Genes (Basel) 2025; 16:191. [PMID: 40004522 PMCID: PMC11855312 DOI: 10.3390/genes16020191] [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: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The experience of prenatal stress results in various physiological disorders due to an alteration of an offspring's methylome and transcriptome. The objective of this study was to determine whether PNS affects DNA methylation (DNAm) and gene expression in the stress axis tissues of mature Brahman cows. Methods: Samples were collected from the paraventricular nucleus (PVN), anterior pituitary (PIT), and adrenal cortex (AC) of 5-year-old Brahman cows that were prenatally exposed to either transportation stress (PNS, n = 6) or were not transported (Control, n = 8). The isolated DNA and RNA samples were, respectively, used for methylation and RNA-Seq analyses. A gene ontology and KEGG pathway enrichment analysis of each data set within each sample tissue was conducted with the DAVID Functional Annotation Tool. Results: The DNAm analysis revealed 3, 64, and 99 hypomethylated and 2, 93, and 90 hypermethylated CpG sites (FDR < 0.15) within the PVN, PIT, and AC, respectively. The RNA-Seq analysis revealed 6, 25, and 5 differentially expressed genes (FDR < 0.15) in the PVN, PIT, and AC, respectively, that were up-regulated in the PNS group relative to the Control group, as well as 24 genes in the PIT that were down-regulated. Based on the enrichment analysis, several developmental and cellular processes, such as maintenance of the actin cytoskeleton, cell motility, signal transduction, neurodevelopment, and synaptic function, were potentially modulated. Conclusions: The methylome and transcriptome were altered in the stress axis tissues of mature cows that had been exposed to prenatal transportation stress. These findings are relevant to understanding how prenatal experiences may affect postnatal neurological functions.
Collapse
Affiliation(s)
- Audrey L. Earnhardt-San
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Texas A&M AgriLife Research Center, Overton, TX 75684, USA
| | - Emilie C. Baker
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Kubra Z. Cilkiz
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Rodolfo C. Cardoso
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Noushin Ghaffari
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Charles R. Long
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Penny K. Riggs
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Ronald D. Randel
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - David G. Riley
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Thomas H. Welsh
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| |
Collapse
|
6
|
Gorecki L, Reznickova E, Krystof V, Rezacova M, Ceckova M, Korabecny J. Strategies for the treatment of acute myeloid leukemia with FLT3 mutations: a patent review. Expert Opin Ther Pat 2025; 35:137-164. [PMID: 39718422 DOI: 10.1080/13543776.2024.2446224] [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: 08/29/2024] [Revised: 11/09/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION Approximately one-third of all AML patients have a mutation in the Fms-like tyrosine kinase 3 (FLT3) gene, which is associated with a poor prognosis in these individuals. The 2017 approval of midostaurin, the first FLT3 inhibitor, spurred extensive development of more potent and selective inhibitors with an improved safety profile. AREAS COVERED This review analyzes patent inventions for the treatment of AML using FLT3 inhibitors, covering developments from the earliest to the most recent, disclosed in 2024. Our search using the global Espacenet database identified numerous compounds with low nanomolar inhibitory concentrations against FLT3-ITD and FLT3-TKD mutants. These compounds have shown promise in preclinical studies. Co-inhibition strategies and combinatorial therapies to overcome resistance and enhance anti-leukemic efficacy are also discussed. EXPERT OPINION Recent patents highlight advances in the field of FLT3 inhibitors with a focus on overcoming resistance, improving selectivity and potency. Future strategies may include third-generation inhibitors such as type III allosteric inhibitors, irreversible inhibitors, or PROTACs. Personalized medicine approaches utilizing genetic profiling to tailor therapies are emphasized. Exploration of novel combination regimens with emerging therapies like CAR T-cell therapy, immune checkpoint inhibitors, and small molecules targeting critical AML pathways is ongoing to further enhance anti-leukemic efficacy.
Collapse
Affiliation(s)
- Lukas Gorecki
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Hradec Kralove, Czech Republic
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Eva Reznickova
- Department of Experimental Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Vladimir Krystof
- Department of Experimental Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Martina Rezacova
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Králové, Charles University, Hradec Kralove, Czech Republic
| | - Martina Ceckova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
7
|
Germini D, Farhat R, Dadon L, Lacroix A, Nemati F, Rebollo A, Decaudin D, Wiels J, Brenner C. A translational study for biomarker identification of PEP-010, a pro-apoptotic peptide restoring apoptosis in cancer models. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167492. [PMID: 39218274 DOI: 10.1016/j.bbadis.2024.167492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/31/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Affiliation(s)
- D Germini
- PEP-Therapy, 111 Avenue de France, 75013 Paris, France.
| | - R Farhat
- PEP-Therapy, 111 Avenue de France, 75013 Paris, France
| | - L Dadon
- PEP-Therapy, 111 Avenue de France, 75013 Paris, France
| | - A Lacroix
- PEP-Therapy, 111 Avenue de France, 75013 Paris, France
| | - F Nemati
- Translational Research Department, Laboratory of Preclinical Investigation, PSL University, Institut Curie, 26 rue d'Ulm, Paris 75248, France
| | - A Rebollo
- Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), INSERM U1267 - CNRS UMR8258, Université de Paris, Faculté de Pharmacie, France
| | - D Decaudin
- Translational Research Department, Laboratory of Preclinical Investigation, PSL University, Institut Curie, 26 rue d'Ulm, Paris 75248, France; Department of Medical Oncology, Institut Curie, 26 rue d'Ulm, Paris 75248, France
| | - J Wiels
- Université Paris-Saclay, CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, 94805 Villejuif, France
| | - C Brenner
- Université Paris-Saclay, CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, 94805 Villejuif, France.
| |
Collapse
|
8
|
Dabbaghi MM, Soleimani Roudi H, Safaei R, Baradaran Rahimi V, Fadaei MR, Askari VR. Unveiling the Mechanism of Protective Effects of Tanshinone as a New Fighter Against Cardiovascular Diseases: A Systematic Review. Cardiovasc Toxicol 2024; 24:1467-1509. [PMID: 39306819 DOI: 10.1007/s12012-024-09921-x] [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: 06/28/2024] [Accepted: 09/08/2024] [Indexed: 11/15/2024]
Abstract
Tanshinone, a natural compound found in the roots of Salvia miltiorrhiza, has been shown to possess various pharmacological properties, including anti-inflammatory, antioxidant, and cardiovascular protective effects. This article aims to review the literature on the cardiovascular protective effects of tanshinone and its underlying mechanisms. Tanshinone has been demonstrated to improve cardiac function, reduce oxidative stress, and inhibit inflammation in various animal models of cardiovascular diseases. Additionally, it has been shown to regulate multiple signaling pathways involved in the pathogenesis of cardiovascular diseases, such as the PI3K/AKT, MAPK, and NF-κB pathways. Clinical studies have also suggested that tanshinone may have therapeutic potential for treating cardiovascular diseases. In conclusion, tanshinone has emerged as a promising natural compound with significant cardiovascular protective effects, and further research is warranted to explore its potential clinical applications.
Collapse
Affiliation(s)
- Mohammad Mahdi Dabbaghi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran
| | - Hesan Soleimani Roudi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran
| | - Rozhan Safaei
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Fadaei
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran.
| |
Collapse
|
9
|
Luo W, Li N, Liu J, Li D, Li Y, Ma Q, Lin C, Lu L, Lin S. Bulk and Single-Cell Transcriptome Analyses Unravel Gene Signatures of Mitochondria-Associated Programmed Cell Death in Diabetic Foot Ulcer. J Cell Mol Med 2024; 28:e70319. [PMID: 39730319 DOI: 10.1111/jcmm.70319] [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: 09/06/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/29/2024] Open
Abstract
Mitochondrial programmed cell death (PCD) plays a critical role in the pathogenesis of diabetic foot ulcers (DFU). In this study, we performed a comprehensive transcriptome analysis to identify potential hub genes and key cell types associated with PCD and mitochondria in DFU. Using intersection analysis of PCD- and mitochondria-related genes, we identified candidate hub genes through protein-protein interaction and random forest analysis. At the single-cell level, key cell types were further validated based on the expression of hub genes. Additionally, we explored the transcription factors (TFs) regulating hub gene expression and the cellular heterogeneity of DFU. Finally, the expression of key hub genes and TFs was validated in clinical specimens. Our results identified BCL2 and LIPT1 as significantly downregulated hub genes in DFU, with Keratinocytes, as the key cell type. Immunohistochemistry confirmed downregulation of BCL2 and LIPT1 in DFU samples (p < 0.05). Additionally, TFs CEBPD and IRF1 were significantly upregulated in DFU, as confirmed by real-time polymerase chain reaction analysis (p < 0.05).
Collapse
Affiliation(s)
- Wenqiang Luo
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Ning Li
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Jin Liu
- Department of Orthopedics, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, P. R. China
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Shanwei, Guangdong, P. R. China
| | - Duoyu Li
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Yongheng Li
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Qing Ma
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Chuangxin Lin
- Department of Orthopedic Surgery, Shantou Central Hospital, Shantou, P. R. China
| | - Liang Lu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Sipeng Lin
- Department of Orthopedics, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, P. R. China
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Shanwei, Guangdong, P. R. China
| |
Collapse
|
10
|
Siraj Y, Aprile D, Alessio N, Peluso G, Di Bernardo G, Galderisi U. IGFBP7 is a key component of the senescence-associated secretory phenotype (SASP) that induces senescence in healthy cells by modulating the insulin, IGF, and activin A pathways. Cell Commun Signal 2024; 22:540. [PMID: 39533382 PMCID: PMC11558980 DOI: 10.1186/s12964-024-01921-2] [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: 08/13/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Senescent cells exert their effects through the release of various factors, collectively referred to as the senescence-associated secretory phenotype (SASP). The SASP can induce senescence in healthy cells (secondary senescence), modulate immune system function, reshape the extracellular matrix, and facilitate cancer progression.Among SASP components, certain factors act as key regulators in the induction of secondary senescence. In this study, we evaluated the role of IGFBP7, a crucial SASP component. Our results demonstrated that ROS-prostaglandin signaling is involved in the release of IGFBP7. Furthermore, neutralizing antibodies targeting IGFBP7 attenuated the SASP's pro-senescence activity. Cells incubated with IGFBP7 also entered a state of senescence.The senescence induced by IGFBP7 appears to be mediated through three primary pathways. First, IGFBP7 can bind to insulin, thereby inhibiting its anti-senescence and pro-growth effects. In addition to this inhibitory effect on the insulin pathway, IGFBP7 may enhance IGFII pro-senescence signaling by promoting its interaction with IGF2R while blocking IGF1R. These activities are dependent on ERK and AKT signaling pathways. Finally, IGFBP7 and Activin A, both of which can induce cellular senescence, appear to regulate and inhibit each other, suggesting a compensatory mechanism to prevent excessive senescence. Notably, our preliminary data indicate that IGFBP7, in addition to blocking Activin A, may interact with its receptors and induce senescence via SMAD pathways.Our findings highlight that IGFBP7, along with other members of the IGFBP family, plays a pivotal role in senescence-related signaling pathways. Therefore, IGFBP7 may serve as a potential target for anti-aging strategies aimed at reducing the burden of senescence on tissues and organs.
Collapse
Affiliation(s)
- Yesuf Siraj
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
- College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
| | - Domenico Aprile
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Nicola Alessio
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | | | - Giovanni Di Bernardo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy.
- Department of Experimental Medicine, Luigi Vanvitelli Campania Univeristy, Via Luigi De Crecchio 7, Napoli, 80138, Italy.
| | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy.
- Genome and Stem Cell Center (GENKÖK), Erciyes University, Kayseri, Turkey.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA, USA.
- Department of Experimental Medicine, Luigi Vanvitelli Campania Univeristy, Via Luigi De Crecchio 7, Napoli, 80138, Italy.
| |
Collapse
|
11
|
Zhang J, Zhou P, Wu T, Zhang L, Kang J, Liao J, Jiang D, Hu Z, Han Z, Zhou B. Metformin combined with cisplatin reduces anticancer activity via ATM/CHK2-dependent upregulation of Rad51 pathway in ovarian cancer. Neoplasia 2024; 57:101037. [PMID: 39142065 PMCID: PMC11379670 DOI: 10.1016/j.neo.2024.101037] [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: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/16/2024]
Abstract
Ovarian cancer (OC) is the deadliest malignancy of the female reproductive system. The standard first-line therapy for OC involves cytoreductive surgical debulking followed by chemotherapy based on platinum and paclitaxel. Despite these treatments, there remains a high rate of tumor recurrence and resistance to platinum. Recent studies have highlighted the potential anti-tumor properties of metformin (met), a traditional diabetes drug. In our study, we investigated the impact of met on the anticancer activities of cisplatin (cDDP) both in vitro and in vivo. Our findings revealed that combining met with cisplatin significantly reduced apoptosis in OC cells, decreased DNA damage, and induced resistance to cDDP. Furthermore, our mechanistic study indicated that the resistance induced by met is primarily driven by the inhibition of the ATM/CHK2 pathway and the upregulation of the Rad51 protein. Using an ATM inhibitor, KU55933, effectively reversed the cisplatin resistance phenotype. In conclusion, our results suggest that met can antagonize the effects of cDDP in specific types of OC cells, leading to a reduction in the chemotherapeutic efficacy of cDDP.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Gynecologic Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei 430071, China; Women and Children's Hospital Afiliated to Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Ping Zhou
- Gynecological Department, Dongguan Maternal and Child Hospital, Dongguan, Guangdong 523000, China
| | - Tiancheng Wu
- Department of Gynecologic Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei 430071, China; Women and Children's Hospital Afiliated to Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Liping Zhang
- Department of Social Medical Development, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Jiaqi Kang
- Department of gynaecology and obstetrics of People's Hospital of Zhongxiang City, Hubei 431900, China
| | - Jing Liao
- Women and Children's Hospital Afiliated to Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China; Department of Gynecology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Daqiong Jiang
- Department of Gynecologic Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei 430071, China; Women and Children's Hospital Afiliated to Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Zheng Hu
- Department of Gynecologic Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei 430071, China; Women and Children's Hospital Afiliated to Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China.
| | - Zhiqiang Han
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Department of Obstetrics and Gynecology, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, China.
| | - Bo Zhou
- Department of Gynecologic Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei 430071, China; Women and Children's Hospital Afiliated to Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China.
| |
Collapse
|
12
|
Chun C, Byun JM, Cha M, Lee H, Choi B, Kim H, Hong S, Lee Y, Park H, Koh Y, Yoon TY. Profiling protein-protein interactions to predict the efficacy of B-cell-lymphoma-2-homology-3 mimetics for acute myeloid leukaemia. Nat Biomed Eng 2024; 8:1379-1395. [PMID: 39025942 PMCID: PMC11584402 DOI: 10.1038/s41551-024-01241-3] [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: 01/15/2023] [Accepted: 06/28/2024] [Indexed: 07/20/2024]
Abstract
B-cell-lymphoma-2 (BCL2) homology-3 (BH3) mimetics are inhibitors of protein-protein interactions (PPIs) that saturate anti-apoptotic proteins in the BCL2 family to induce apoptosis in cancer cells. Despite the success of the BH3-mimetic ABT-199 for the treatment of haematological malignancies, only a fraction of patients respond to the drug and most patients eventually develop resistance to it. Here we show that the efficacy of ABT-199 can be predicted by profiling the rewired status of the PPI network of the BCL2 family via single-molecule pull-down and co-immunoprecipitation to quantify more than 20 types of PPI from a total of only 1.2 × 106 cells per sample. By comparing the obtained multidimensional data with BH3-mimetic efficacies determined ex vivo, we constructed a model for predicting the efficacy of ABT-199 that designates two complexes of the BCL2 protein family as the primary mediators of drug effectiveness and resistance, and applied it to prospectively assist therapeutic decision-making for patients with acute myeloid leukaemia. The characterization of PPI complexes in clinical specimens opens up opportunities for individualized protein-complex-targeting therapies.
Collapse
Affiliation(s)
- Changju Chun
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
| | - Ja Min Byun
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Minkwon Cha
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
- Department of Physics, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Hongwon Lee
- Department of Biomarker Discovery, PROTEINA Co., Ltd, Seoul, South Korea
| | - Byungsan Choi
- Department of Biomarker Discovery, PROTEINA Co., Ltd, Seoul, South Korea
| | - Hyunwoo Kim
- Department of Biomarker Discovery, PROTEINA Co., Ltd, Seoul, South Korea
| | - Saem Hong
- Department of Biomarker Discovery, PROTEINA Co., Ltd, Seoul, South Korea
| | - Yunseo Lee
- Department of Biomarker Discovery, PROTEINA Co., Ltd, Seoul, South Korea
| | - Hayoung Park
- Department of Biomarker Discovery, PROTEINA Co., Ltd, Seoul, South Korea
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea.
| | - Tae-Young Yoon
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, Seoul, South Korea.
- Department of Biomarker Discovery, PROTEINA Co., Ltd, Seoul, South Korea.
| |
Collapse
|
13
|
Ishii T, Mori-Kobayashi K, Nakamura S, Ohkura S, Matsuyama S. Carnosine supplementation in cryopreservation solution improved frozen-thawed bovine embryo viability. J Reprod Dev 2024; 70:279-285. [PMID: 39010149 PMCID: PMC11461515 DOI: 10.1262/jrd.2023-071] [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: 08/10/2023] [Accepted: 06/14/2024] [Indexed: 07/17/2024] Open
Abstract
Cryopreservation adversely affects embryo quality and viability in vitro. We investigated the effects of cryopreservation solutions supplemented with the antioxidant carnosine on frozen-thawed bovine embryo viability. Bovine blastocysts were produced in vitro and cryopreserved using slow freezing. The rates of re-expanded and hatched blastocysts in the 50 μg/ml carnosine-supplemented group at 4, 24, and 48 h after thawing were higher than those in the control (P < 0.05) group. In frozen-thawed embryos, cryopreservation solution supplemented with carnosine (50 μg/ml) significantly reduced reactive oxygen species (ROS) production (P < 0.05), decreased TUNEL-positive apoptotic cells (P < 0.05), and increased the mRNA expression of BCL2 (P < 0.05), an apoptosis suppressor gene. The expression of translocase of outer mitochondrial membrane 20 (TOMM20), which is involved in protein mitochondrial transport, in the carnosine (50 μg/ml)-treated embryos was significantly higher than that in the control group (P < 0.05). ATP production in frozen-thawed embryos in the 50 μg/ml carnosine-supplemented group was significantly higher than that in the control group (P < 0.05), however no significant difference in the total number of cells per embryo among the groups was observed. These results suggest that supplementing the cryopreservation solution with carnosine can improve the viability of frozen-thawed bovine embryos by reducing oxidative damage.
Collapse
Affiliation(s)
- Toshimichi Ishii
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
- Mie Prefectural Livestock Research Center, Mie 515-2324, Japan
| | | | - Sho Nakamura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Satoshi Ohkura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Shuichi Matsuyama
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| |
Collapse
|
14
|
Kumar Mahanta S, Pratikshya Nayak P, Muduli K, Elangovan S, Paramasivan SS, Kumar Mallick P, Kumar Mohapatra S, Kumar Panda S. Optimisation ofLevilactobacillus brevis-fermented finger millet (Eleusine coracana) and evaluation of its effects on cancer cells (HCT116 and MDA-MB-231). Methods 2024; 229:30-40. [PMID: 38880434 DOI: 10.1016/j.ymeth.2024.06.002] [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: 05/02/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024] Open
Abstract
The objective of this study was to optimise the millet formulation using Levilactobacillus brevis and to evaluate its anticarcinogenic potential in vitro. The formula was developed in the course of the fermentation of finger millet (Eleusine coracana) using L. brevis MTTC 4460 and optimised by response surface methodology and validation by artificial neural networking (ANN). The optimised millet formulation could be obtained using 2 % of bacterial inoculum, 2 % of glucose, and a fermentation duration of 3.3 days with a yield of 5.98 mg/mL lactic acid and 3.38 log10 (CFU/mL) viable L. brevis with overall desirability value of 1. The fermented millet formulation exhibited antiproliferative and antimigratory effects on MDA-MB-231 and HCT116 cancer cell lines. In addition, the outcomes observed in western blot analysis revealed that the formulation elicited apoptotic responses mediated by the Bcl-2 family of proteins in MDA-MB-231 and HCT116 cell lines while demonstrating no discernible impact on HEK293 normal cells.
Collapse
Affiliation(s)
- Sachin Kumar Mahanta
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Priyadarshini Pratikshya Nayak
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Kartik Muduli
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Selvakumar Elangovan
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Sethuraman Sivakumar Paramasivan
- Section of Extension and Social Sciences, ICAR - Central Tuber Crops Research Institute, Thiruvananthapuram - 695017, Kerala, India
| | - Pradeep Kumar Mallick
- School of Computer Engineering, Campus 15, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | | | - Sandeep Kumar Panda
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India.
| |
Collapse
|
15
|
Dzhemileva LU, D'yakonov VA, Egorova KS, Ananikov VP. Mechanisms of cytotoxicity in six classes of ionic liquids: Evaluating cell cycle impact and genotoxic and apoptotic effects. CHEMOSPHERE 2024; 364:142964. [PMID: 39074667 DOI: 10.1016/j.chemosphere.2024.142964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/03/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
Ionic liquids (ILs), earlier praised for their eco-friendliness, have emerged as key chemicals in advancing green chemistry, catalysis, solvent development, and more. However, the discovery of their notable toxicity has led to a controversial reputation of ILs and has shifted the research landscape towards understanding their biological impacts. The present study examines the mechanism of cytotoxicity of 32 ILs across six classes, highlighting their effects on the cell cycle of the Jurkat cell line. Focusing on five ILs with pronounced cytotoxicity, we uncover their genotoxic effects and their role in inducing apoptosis. Our findings suggest intricate interplay between the extrinsic and intrinsic apoptotic pathways at different time points after exposure to ILs. Moreover, the ILs studied displayed marked genotoxicity, likely stemming from the accumulation of double-strand DNA breaks in the Jurkat cells. This investigation offers a comprehensive view on interactions of ILs with eukaryotic cells, thereby providing new guidelines for developing safer pharmaceutical and industrial applications of these chemicals. The results not only broaden and enhance the previous perceptions but also open new avenues in research, emphasizing the dual potential of ILs in innovation and safety, and marking a significant step towards integrating chemical innovations with biological safety.
Collapse
Affiliation(s)
- Lilya U Dzhemileva
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Vladimir A D'yakonov
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Ksenia S Egorova
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Valentine P Ananikov
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
16
|
Tam KP, Xie J, Au-Yeung RKH, Chiang AKS. Combination of bortezomib and venetoclax targets the pro-survival function of LMP-1 and EBNA-3C of Epstein-Barr virus in spontaneous lymphoblastoid cell lines. PLoS Pathog 2024; 20:e1012250. [PMID: 39325843 PMCID: PMC11481030 DOI: 10.1371/journal.ppat.1012250] [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: 05/10/2024] [Revised: 10/08/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Epstein-Barr virus (EBV) manipulates the ubiquitin-proteasome system and regulators of Bcl-2 family to enable the persistence of the virus and survival of the host cells through the expression of viral proteins in distinct latency patterns. We postulate that the combination of bortezomib (proteasome inhibitor) and venetoclax (Bcl-2 inhibitor) [bort/venetoclax] will cause synergistic killing of post-transplant lymphoproliferative disorder (PTLD) through targeting the pro-survival function of latent viral proteins such as latent membrane protein-1 (LMP-1) and EBV nuclear antigen-3C (EBNA-3C). Bort/venetoclax could synergistically kill spontaneous lymphoblastoid cell lines (sLCLs) derived from patients with PTLD and EBV-associated hemophagocytic lymphohistiocytosis by inducing DNA damage response, apoptosis and G1-S cell cycle arrest in a ROS-dependent manner. Bortezomib potently induced the expression of Noxa, a pro-apoptotic initiator and when combined with venetoclax, inhibited Mcl-1 and Bcl-2 simultaneously. Bortezomib prevented LMP-1 induced proteasomal degradation of IκBα leading to the suppression of the NF-κB signaling pathway. Bortezomib also rescued Bcl-6 from EBNA-3C mediated proteasomal degradation thus maintaining the repression of cyclin D1 and Bcl-2 causing G1-S arrest and apoptosis. Concurrently, venetoclax inhibited Bcl-2 upregulated by either LMP-1 or EBNA-3C. Bort/venetoclax decreased the expression of phosphorylated p65 and Bcl-2 at serine 70 thereby suppressing the NF-κB signaling pathway and promoting apoptosis, respectively. These data corroborated the marked suppression of the growth of xenograft of sLCL in SCID mice (p<0.001). Taken together, the combination of bortezomib and venetoclax targets the pro-survival function of LMP-1 and EBNA-3C of Epstein-Barr virus in spontaneous lymphoblastoid cell lines.
Collapse
Affiliation(s)
- Kam Pui Tam
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jia Xie
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Rex Kwok Him Au-Yeung
- Department of Pathology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Alan K. S. Chiang
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
17
|
Yang Z, Deng X, Wen D, Sun L, An R, Xu J. Identification of RCAN1's role in hepatocellular carcinoma using single-cell analysis. BMC Cancer 2024; 24:1056. [PMID: 39192218 PMCID: PMC11348566 DOI: 10.1186/s12885-024-12807-4] [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: 01/31/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND The regulator of calcineurin 1 (RCAN1) is expressed in multiple organs, including the heart, liver, brain, and kidney, and is closely linked to the pathogenesis of cardiovascular diseases, Down syndrome, and Alzheimer's disease. It is also implicated in the development of various organ tumors; however, its potential role in hepatocellular carcinoma (HCC) remains poorly understood. Therefore, the objective of this study was to investigate the potential mechanisms of RCAN1 in HCC through bioinformatics analysis. METHODS We conducted a joint analysis based on the NCBI and TCGA databases, integrating both bulk transcriptome and single-cell analyses to examine the principal biological functions of RCAN1 in HCC, as well as its roles related to phenotype, metabolism, and cell communication. Subsequently, an RCAN1-overexpressing cell line was established, and the effects of RCAN1 on tumor cells were validated through in vitro experiments. Moreover, we endeavored to identify potential related drugs using molecular docking and molecular dynamics simulations. RESULTS The expression of RCAN1 was found to be downregulated in 19 types of cancer tissues and upregulated in 11 types of cancer tissues. Higher levels of RCAN1 expression were associated with improved patient survival. RCAN1 was predominantly expressed in hepatocytes, macrophages, endothelial cells, and monocytes, and its high expression not only closely correlated with the distribution of cells related to the HCC phenotype but also with the distribution of HCC cells themselves. Additionally, Rcan1 may directly or indirectly participate in metabolic pathways such as alanine, aspartate, and glutamate metabolism, as well as butanoate metabolism, thereby influencing tumor cell proliferation and migration. In vitro experiments confirmed that RCAN1 overexpression promoted apoptosis while inhibiting proliferation and invasion of HCC cells. Through molecular docking of 1615 drugs, we screened brompheniramine as a potential target drug and verified our results by molecular dynamics. CONCLUSION In this study, we revealed the relationship between RCAN1 and HCC through bioinformatics methods, verified that RCAN1 can affect the progress of the disease through experiments, and finally identified potential therapeutic drugs through drug molecular docking and molecular dynamics.
Collapse
Affiliation(s)
- Ziqi Yang
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Interventional Surgery Center, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiwei Deng
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Interventional Surgery Center, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Oncology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Didi Wen
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lijun Sun
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Interventional Surgery Center, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Rui An
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
- Department of Interventional Surgery Center, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Jian Xu
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
- Department of Interventional Surgery Center, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
18
|
Jiang Y, Rex DA, Schuster D, Neely BA, Rosano GL, Volkmar N, Momenzadeh A, Peters-Clarke TM, Egbert SB, Kreimer S, Doud EH, Crook OM, Yadav AK, Vanuopadath M, Hegeman AD, Mayta M, Duboff AG, Riley NM, Moritz RL, Meyer JG. Comprehensive Overview of Bottom-Up Proteomics Using Mass Spectrometry. ACS MEASUREMENT SCIENCE AU 2024; 4:338-417. [PMID: 39193565 PMCID: PMC11348894 DOI: 10.1021/acsmeasuresciau.3c00068] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 08/29/2024]
Abstract
Proteomics is the large scale study of protein structure and function from biological systems through protein identification and quantification. "Shotgun proteomics" or "bottom-up proteomics" is the prevailing strategy, in which proteins are hydrolyzed into peptides that are analyzed by mass spectrometry. Proteomics studies can be applied to diverse studies ranging from simple protein identification to studies of proteoforms, protein-protein interactions, protein structural alterations, absolute and relative protein quantification, post-translational modifications, and protein stability. To enable this range of different experiments, there are diverse strategies for proteome analysis. The nuances of how proteomic workflows differ may be challenging to understand for new practitioners. Here, we provide a comprehensive overview of different proteomics methods. We cover from biochemistry basics and protein extraction to biological interpretation and orthogonal validation. We expect this Review will serve as a handbook for researchers who are new to the field of bottom-up proteomics.
Collapse
Affiliation(s)
- Yuming Jiang
- Department
of Computational Biomedicine, Cedars Sinai
Medical Center, Los Angeles, California 90048, United States
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Devasahayam Arokia
Balaya Rex
- Center for
Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Dina Schuster
- Department
of Biology, Institute of Molecular Systems
Biology, ETH Zurich, Zurich 8093, Switzerland
- Department
of Biology, Institute of Molecular Biology
and Biophysics, ETH Zurich, Zurich 8093, Switzerland
- Laboratory
of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, Villigen 5232, Switzerland
| | - Benjamin A. Neely
- Chemical
Sciences Division, National Institute of
Standards and Technology, NIST, Charleston, South Carolina 29412, United States
| | - Germán L. Rosano
- Mass
Spectrometry
Unit, Institute of Molecular and Cellular
Biology of Rosario, Rosario, 2000 Argentina
| | - Norbert Volkmar
- Department
of Biology, Institute of Molecular Systems
Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Amanda Momenzadeh
- Department
of Computational Biomedicine, Cedars Sinai
Medical Center, Los Angeles, California 90048, United States
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Trenton M. Peters-Clarke
- Department
of Pharmaceutical Chemistry, University
of California—San Francisco, San Francisco, California, 94158, United States
| | - Susan B. Egbert
- Department
of Chemistry, University of Manitoba, Winnipeg, Manitoba, R3T 2N2 Canada
| | - Simion Kreimer
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Emma H. Doud
- Center
for Proteome Analysis, Indiana University
School of Medicine, Indianapolis, Indiana, 46202-3082, United States
| | - Oliver M. Crook
- Oxford
Protein Informatics Group, Department of Statistics, University of Oxford, Oxford OX1 3LB, United
Kingdom
| | - Amit Kumar Yadav
- Translational
Health Science and Technology Institute, NCR Biotech Science Cluster 3rd Milestone Faridabad-Gurgaon
Expressway, Faridabad, Haryana 121001, India
| | | | - Adrian D. Hegeman
- Departments
of Horticultural Science and Plant and Microbial Biology, University of Minnesota, Twin Cities, Minnesota 55108, United States
| | - Martín
L. Mayta
- School
of Medicine and Health Sciences, Center for Health Sciences Research, Universidad Adventista del Plata, Libertador San Martin 3103, Argentina
- Molecular
Biology Department, School of Pharmacy and Biochemistry, Universidad Nacional de Rosario, Rosario 2000, Argentina
| | - Anna G. Duboff
- Department
of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Nicholas M. Riley
- Department
of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Robert L. Moritz
- Institute
for Systems biology, Seattle, Washington 98109, United States
| | - Jesse G. Meyer
- Department
of Computational Biomedicine, Cedars Sinai
Medical Center, Los Angeles, California 90048, United States
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| |
Collapse
|
19
|
Huang SY, Gong S, Zhao Y, Ye ML, Li JY, He QM, Qiao H, Tan XR, Wang JY, Liang YL, Huang SW, He SW, Li YQ, Xu S, Li YQ, Liu N. PJA1-mediated suppression of pyroptosis as a driver of docetaxel resistance in nasopharyngeal carcinoma. Nat Commun 2024; 15:5300. [PMID: 38906860 PMCID: PMC11192944 DOI: 10.1038/s41467-024-49675-2] [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: 09/27/2023] [Accepted: 06/17/2024] [Indexed: 06/23/2024] Open
Abstract
Chemoresistance is a main reason for treatment failure in patients with nasopharyngeal carcinoma, but the exact regulatory mechanism underlying chemoresistance in nasopharyngeal carcinoma remains to be elucidated. Here, we identify PJA1 as a key E3 ubiquitin ligase involved in nasopharyngeal carcinoma chemoresistance that is highly expressed in nasopharyngeal carcinoma patients with nonresponse to docetaxel-cisplatin-5-fluorouracil induction chemotherapy. We find that PJA1 facilitates docetaxel resistance by inhibiting GSDME-mediated pyroptosis in nasopharyngeal carcinoma cells. Mechanistically, PJA1 promotes the degradation of the mitochondrial protein PGAM5 by increasing its K48-linked ubiquitination at K88, which further facilitates DRP1 phosphorylation at S637 and reduced mitochondrial reactive oxygen species production, resulting in suppression of GSDME-mediated pyroptosis and the antitumour immune response. PGAM5 knockdown fully restores the docetaxel sensitization effect of PJA1 knockdown. Moreover, pharmacological targeting of PJA1 with the small molecule inhibitor RTA402 enhances the docetaxel sensitivity of nasopharyngeal carcinoma in vitro and in vivo. Clinically, high PJA1 expression indicates inferior survival and poor clinical efficacy of TPF IC in nasopharyngeal carcinoma patients. Our study emphasizes the essential role of E3 ligases in regulating chemoresistance and provides therapeutic strategies for nasopharyngeal carcinoma based on targeting the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Sheng-Yan Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Sha Gong
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Yin Zhao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Ming-Liang Ye
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Jun-Yan Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Qing-Mei He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Han Qiao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Xi-Rong Tan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Jing-Yun Wang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Ye-Lin Liang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Sai-Wei Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Shi-Wei He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Ying-Qin Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Sha Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Ying-Qing Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
| | - Na Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
| |
Collapse
|
20
|
Kesarwani M, Kincaid Z, Azhar M, Azam M. Enhanced MAPK signaling induced by CSF3R mutants confers dependence to DUSP1 for leukemic transformation. Blood Adv 2024; 8:2765-2776. [PMID: 38531054 PMCID: PMC11176961 DOI: 10.1182/bloodadvances.2023010830] [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: 05/26/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/28/2024] Open
Abstract
ABSTRACT Elevated MAPK and the JAK-STAT signaling play pivotal roles in the pathogenesis of chronic neutrophilic leukemia and atypical chronic myeloid leukemia. Although inhibitors targeting these pathways effectively suppress the diseases, they fall short in providing enduring remission, largely attributed to the cytostatic nature of these drugs. Even combinations of these drugs are ineffective in achieving sustained remission. Enhanced MAPK signaling besides promoting proliferation and survival triggers a proapoptotic response. Consequently, malignancies reliant on elevated MAPK signaling use MAPK feedback regulators to intricately modulate the signaling output, prioritizing proliferation and survival while dampening the apoptotic stimuli. Herein, we demonstrate that enhanced MAPK signaling in granulocyte colony-stimulating factor 3 receptor (CSF3R)-driven leukemia upregulates the expression of dual specificity phosphatase 1 (DUSP1) to suppress the apoptotic stimuli crucial for leukemogenesis. Consequently, genetic deletion of Dusp1 in mice conferred synthetic lethality to CSF3R-induced leukemia. Mechanistically, DUSP1 depletion in leukemic context causes activation of JNK1/2 that results in induced expression of BIM and P53 while suppressing the expression of BCL2 that selectively triggers apoptotic response in leukemic cells. Pharmacological inhibition of DUSP1 by BCI (a DUSP1 inhibitor) alone lacked antileukemic activity due to ERK1/2 rebound caused by off-target inhibition of DUSP6. Consequently, a combination of BCI with a MEK inhibitor successfully cured CSF3R-induced leukemia in a preclinical mouse model. Our findings underscore the pivotal role of DUSP1 in leukemic transformation driven by enhanced MAPK signaling and advocate for the development of a selective DUSP1 inhibitor for curative treatment outcomes.
Collapse
Affiliation(s)
- Meenu Kesarwani
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
| | - Zachary Kincaid
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
| | - Mohammad Azhar
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
| | - Mohammad Azam
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
21
|
Ahator SD, Hegstad K, Lentz CS, Johannessen M. Deciphering Staphylococcus aureus-host dynamics using dual activity-based protein profiling of ATP-interacting proteins. mSystems 2024; 9:e0017924. [PMID: 38656122 PMCID: PMC11097646 DOI: 10.1128/msystems.00179-24] [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: 02/06/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
The utilization of ATP within cells plays a fundamental role in cellular processes that are essential for the regulation of host-pathogen dynamics and the subsequent immune response. This study focuses on ATP-binding proteins to dissect the complex interplay between Staphylococcus aureus and human cells, particularly macrophages (THP-1) and keratinocytes (HaCaT), during an intracellular infection. A snapshot of the various protein activity and function is provided using a desthiobiotin-ATP probe, which targets ATP-interacting proteins. In S. aureus, we observe enrichment in pathways required for nutrient acquisition, biosynthesis and metabolism of amino acids, and energy metabolism when located inside human cells. Additionally, the direct profiling of the protein activity revealed specific adaptations of S. aureus to the keratinocytes and macrophages. Mapping the differentially activated proteins to biochemical pathways in the human cells with intracellular bacteria revealed cell-type-specific adaptations to bacterial challenges where THP-1 cells prioritized immune defenses, autophagic cell death, and inflammation. In contrast, HaCaT cells emphasized barrier integrity and immune activation. We also observe bacterial modulation of host processes and metabolic shifts. These findings offer valuable insights into the dynamics of S. aureus-host cell interactions, shedding light on modulating host immune responses to S. aureus, which could involve developing immunomodulatory therapies. IMPORTANCE This study uses a chemoproteomic approach to target active ATP-interacting proteins and examines the dynamic proteomic interactions between Staphylococcus aureus and human cell lines THP-1 and HaCaT. It uncovers the distinct responses of macrophages and keratinocytes during bacterial infection. S. aureus demonstrated a tailored response to the intracellular environment of each cell type and adaptation during exposure to professional and non-professional phagocytes. It also highlights strategies employed by S. aureus to persist within host cells. This study offers significant insights into the human cell response to S. aureus infection, illuminating the complex proteomic shifts that underlie the defense mechanisms of macrophages and keratinocytes. Notably, the study underscores the nuanced interplay between the host's metabolic reprogramming and immune strategy, suggesting potential therapeutic targets for enhancing host defense and inhibiting bacterial survival. The findings enhance our understanding of host-pathogen interactions and can inform the development of targeted therapies against S. aureus infections.
Collapse
Affiliation(s)
- Stephen Dela Ahator
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Kristin Hegstad
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Christian S. Lentz
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Mona Johannessen
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
22
|
Yang X, She X, Zhao Z, Ren J, Wang P, Dong H, Zhao QS, Liu J. In vitro and vivo anti-tumor activity and mechanisms of the new cryptotanshinone derivative 11 against hepatocellular carcinoma. Eur J Pharmacol 2024; 971:176522. [PMID: 38522640 DOI: 10.1016/j.ejphar.2024.176522] [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: 10/22/2023] [Revised: 03/06/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Global burden of hepatocellular carcinoma (HCC) is increasing. Chemotherapy and immunotherapy are the prevailing options for therapy. Developing new therapeutic strategies for HCC patients is still highly desirable. Recent studies demonstrate that cryptotanshinone is capable of inhibiting tumor growth in HCC and induces antitumor immunity in vitro. In our previous research, we discovered a new cryptotanshinone derivative 11 as an effective immunoregulatory enzyme indoleamine 2, 3-dioxygenase 1 (IDO1) inhibitor. This study aims to evaluate its in vitro and in vivo antitumor activity against hepatocellular carcinoma. 11 displayed robust anti-proliferative activity against HCC cell lines and promoted apoptosis of HCC cell line through the mitochondrial-mediated apoptotic pathway. In H22 tumor-bearing mice models, 11 exhibited significant in vivo anti-tumor activity with different administration routes. And no obvious toxicity was observed. RNA-seq analysis demonstrated the differential expressed genes and alteration of key pathways associated with immune responses after administration of 11. Up-regulation of anti-tumor cytokines and down-regulation of cytokines that promote tumor growth were indicated and further validated. Our study demonstrates that 11 exhibits promising anti-tumor activity both in vitro and in vivo against hepatocellular carcinoma cancer. It is a lead compound for HCC immunotherapy and is worthy for further development.
Collapse
Affiliation(s)
- Xinni Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Xianlan She
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Zhishuang Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jian Ren
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Peiying Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Haoqi Dong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qin-Shi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Jiangxin Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| |
Collapse
|
23
|
Ren X, Wang X, Song H, Zhang C, Yuan J, He J, Li J, Wang Z. Long non-coding RNA LINC01554 overexpression suppresses viability, migration, and invasion of liver cancer cells through regulating miR-148b-3p/EIF4E3. Heliyon 2024; 10:e27319. [PMID: 38501022 PMCID: PMC10945188 DOI: 10.1016/j.heliyon.2024.e27319] [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: 09/05/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024] Open
Abstract
Background Long non-coding RNAs (lncRNAs) can be severed as competing endogenous RNAs (ceRNAs) to regulate target genes or mRNAs via sponging microRNAs (miRNAs). This study explored the effect of LINC01554 on liver cancer cells through the ceRNA mechanism. Methods Five significantly down-regulated lncRNAs were selected for further verification, and then through bioinformatics, interactive miRNAs and mRNAs of lncRNAs were identified. The relationship between LINC01554, miR-148b-3p and EIF4E3 was detected by the dual luciferase reporter gene assay. Afterwards, HCCLM3 cells were transfected with pCDH-LINC01554, miR-148b-3p inhibitor and miR-148b-3p mimics. Cell viability, apoptosis, migration and invasion were measured by Cell Counting Kit-8, flow cytometer, and Transwell assays. Real-time quantitative PCR (RT-qPCR) and Western blot were used to measure the expressions of related genes and proteins. Results LINC01554 was significantly down-regulated in the liver cancer cell lines, and was expressed in the cytoplasm of HCCLM3 cells. LINC01554 overexpression inhibited proliferation, migration, and invasion of HCCLM3 cells, and promote their apoptosis (P < 0.05). Besides, LINC01554 overexpression also significantly increased the levels of BAX, BCL2/BAX, P53, cleaved-Caspase3, TIMP3, E-cadherin and EIF4E3 (P < 0.05). Through bioinformatics and dual-luciferase reporter gene assay, LINC01554, miR-148b-3p and EIF4E3 were proved to interact with each other. Furthermore, the effects of miR-148b-3p knockdown on HCCLM3 cells were similar with those of LINC01554 overexpression, and miR-148b-3p mimics could reverse the changes of cell viability, apoptosis, migration, and invasion induced by LINC01554 overexpression. Conclusions LINC01554 overexpression could suppress the growth and metastasis of HCCLM3 cells via miR-148b-3p/EIF4E3.
Collapse
Affiliation(s)
- Xiaojing Ren
- Radiological & Environment Medicine Dept, China Institute for Radiation Protection, Taiyuan, 030032, China
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Xiaoxiao Wang
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Huangqin Song
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Chao Zhang
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Junlong Yuan
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Jiefeng He
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Jianguo Li
- Radiological & Environment Medicine Dept, China Institute for Radiation Protection, Taiyuan, 030032, China
| | - Zhuangqiang Wang
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| |
Collapse
|
24
|
Zhou H, Sun X, Dai Y, Wang X, Dai Z, Li X. 14-3-3-η interacts with BCL-2 to protect human endothelial progenitor cells from ox-LDL-triggered damage. Cell Biol Int 2024; 48:290-299. [PMID: 38100125 DOI: 10.1002/cbin.12105] [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: 05/26/2023] [Revised: 10/31/2023] [Accepted: 11/18/2023] [Indexed: 02/15/2024]
Abstract
Oxidized low-density lipoprotein (ox-LDL) causes dysfunction of endothelial progenitor cells (EPCs), and we recently reported that 14-3-3-η can attenuate the damage triggered by ox-LDL in EPCs. However, the molecular mechanisms by which 14-3-3-η protects EPCs from the damage caused by ox-LDL are not fully understood. In this study, we observed that the expression of 14-3-3-η and BCL-2 were downregulated in ox-LDL-treated EPCs. Overexpression of 14-3-3-η in ox-LDL-treated EPC significantly increased BCL-2 level, while knockdown of BCL-2 reduced 14-3-3-η expression and mitigated the protective effect of 14-3-3-η on EPCs. In addition, we discovered that 14-3-3-η colocalizes and interacts with BCL-2 in EPCs. Taken together, these data suggest that 14-3-3-η protects EPCs from ox-LDL-induced damage by its interaction with BCL-2.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaopei Sun
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yi Dai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaotong Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhihong Dai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiuli Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
25
|
Mendez-Callejas G, Piñeros-Avila M, Celis CA, Torrenegra R, Espinosa-Benitez A, Pestana-Nobles R, Yosa-Reyes J. Natural 2',4-Dihydroxy-4',6'-dimethoxy Chalcone Isolated from Chromolaena tacotana Inhibits Breast Cancer Cell Growth through Autophagy and Mitochondrial Apoptosis. PLANTS (BASEL, SWITZERLAND) 2024; 13:570. [PMID: 38475417 DOI: 10.3390/plants13050570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 03/14/2024]
Abstract
Breast cancer (BC) is one of the most common cancers among women. Effective treatment requires precise tailoring to the genetic makeup of the cancer for improved efficacy. Numerous research studies have concentrated on natural compounds and their anti-breast cancer properties to improve the existing treatment options. Chromolaena tacotana (Klatt) R.M. King and H. Rob (Ch. tacotana) is a notable source of bioactive hydroxy-methylated flavonoids. However, the specific anti-BC mechanisms of these flavonoids, particularly those present in the plant's inflorescences, remain partly undefined. This study focuses on assessing a chalcone derivative extracted from Ch. tacotana inflorescences for its potential to concurrently activate regulated autophagy and intrinsic apoptosis in luminal A and triple-negative BC cells. We determined the chemical composition of the chalcone using ultraviolet (UV) and nuclear magnetic resonance (NMR) spectroscopy. Its selective cytotoxicity against BC cell lines was assessed using the MTT assay. Flow cytometry and Western blot analysis were employed to examine the modulation of proteins governing autophagy and the intrinsic apoptosis pathway. Additionally, in silico simulations were conducted to predict interactions between chalcone and various anti-apoptotic proteins, including the mTOR protein. Chalcone was identified as 2',4-dihydroxy-4',6'-dimethoxy-chalcone (DDC). This compound demonstrated a selective inhibition of BC cell proliferation and triggered autophagy and intrinsic apoptosis. It induced cell cycle arrest in the G0/G1 phase and altered mitochondrial outer membrane potential (∆ψm). The study detected the activation of autophagic LC3-II and mitochondrial pro-apoptotic proteins in both BC cell lines. The regulation of Bcl-XL and Bcl-2 proteins varied according to the BC subtype, yet they showed promising molecular interactions with DDC. Among the examined pro-survival proteins, mTOR and Mcl-1 exhibited the most favorable binding energies and were downregulated in BC cell lines. Further research is needed to fully understand the molecular dynamics involved in the activation and interaction of autophagy and apoptosis pathways in cancer cells in response to potential anticancer agents, like the hydroxy-methylated flavonoids from Ch. tacotana.
Collapse
Affiliation(s)
- Gina Mendez-Callejas
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada (GIBGA), Laboratorio de Biología Celular y Molecular, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A.), Calle 222 #55-37, Bogotá 111166, Colombia
| | - Marco Piñeros-Avila
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada (GIBGA), Laboratorio de Biología Celular y Molecular, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A.), Calle 222 #55-37, Bogotá 111166, Colombia
| | - Crispin A Celis
- Grupo de Investigación en Fitoquímica (GIFUJ), Departamento de Química, Facultad de Ciencias, Pontificia Universidad Javeriana, Cra. 7 #40-62, Bogotá 111321, Colombia
| | - Ruben Torrenegra
- Grupo de Investigación en Productos Naturales de la U.D.C.A. (PRONAUDCA), Laboratorio de Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A.), Calle 222 #55-37, Bogotá 111166, Colombia
| | - Anderson Espinosa-Benitez
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada (GIBGA), Laboratorio de Biología Celular y Molecular, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A.), Calle 222 #55-37, Bogotá 111166, Colombia
| | - Roberto Pestana-Nobles
- Grupo de Investigación en Ciencias Exactas, Física y Naturales Aplicadas, Laboratorio de Simulación Molecular y Bioinformática, Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Carrera 59 #59-65, Barranquilla 080002, Colombia
| | - Juvenal Yosa-Reyes
- Grupo de Investigación en Ciencias Exactas, Física y Naturales Aplicadas, Laboratorio de Simulación Molecular y Bioinformática, Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Carrera 59 #59-65, Barranquilla 080002, Colombia
| |
Collapse
|
26
|
Sun B, Lin L, Yao T, Yao J, Zhang G, Li Y, Li C. Jingfang Granule mitigates Coxsackievirus B3-induced myocardial damage by modulating mucolipin 1 expression. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117396. [PMID: 37951374 DOI: 10.1016/j.jep.2023.117396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/25/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jingfang Granules (JFG) originate from the traditional herbal formula Jingfang Baidu powder. It has the effects of inducing sweating and dispelling wind. It is a classic medication used for treating external pathogenic factors and viral diseases. However, the therapeutic mechanism of JFG for viral myocarditis needs further clarification. AIM OF THE STUDY This study aimed to explore the therapeutic efficacy of JFG on coxsackievirus B3-induced viral myocarditis (VMC), along with the elucidation of its underlying mechanisms. MATERIALS AND METHODS C57 BL/6JNifdc mice were divided randomly into several groups: control, model, Jingfang Granule groups (0.23, 0.46, and 0.69 g/20g, respectively), and a positive group (oseltamivir, 19.33 mg/kg). Following the establishment of the VMC model, the mice underwent an 8 -week treatment regimen. Pathological alterations in cardiac tissues and inflammatory protein expression were monitored. Differential gene analysis was conducted utilizing transcriptomic techniques. The differential gene mucolipin 1 (Mcoln1) was knocked down by transfection with siRNA in H9C2 cell, and investigative techniques such as immunoblotting, qRT-PCR, immunofluorescence, JC-1 staining, reactive oxygen species (ROS) detection, and mitochondrial stress testing were employed to examine its mechanism of action. RESULTS JFG significantly mitigates the pathological damage observed in the cardiac tissues of CVB3-induced VMC mice and attenuates the expression of inflammatory genes. Subsequently, differentially expressed genes are identified through transcriptomic analysis and validated via PCR. Among these, the upregulation of Mcoln1 promotes autophagy, facilitating the clearance of damaged mitochondria and excessive ROS. This has been substantiated through in vitro experiments. Excessive ROS precipitates a reduction in mitochondrial membrane potential, instigating cell apoptosis. In accordance with TUNEL staining results, JFG acts to inhibit cell apoptosis. To ascertain whether Mcoln1 is a crucial target for JFG in treating VMC, Mcoln1 was suppressed in H9C2 cells. The suppression of Mcoln1 hinders the elevation in autophagy levels post-JFG treatment, obstructs the enhancement of mitochondrial function, and impedes the clearance of ROS. Furthermore, the inhibitory effect of JFG on cell apoptosis is attenuated. CONCLUSION The research findings indicate that JFG has a protective effect on CVB3-induced H9C2 cell injury. JFG may exert its effects in VMC treatment by enhancing autophagy to suppress cell apoptosis through the mitochondrial pathway, thereby counteracting cell damage.
Collapse
Affiliation(s)
- Bowen Sun
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Lin Lin
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Tian Yao
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jingchun Yao
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, 276006, China
| | - Guimin Zhang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, 276006, China
| | - Yunlun Li
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China.
| | - Chao Li
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
27
|
Chen L, Li X, Wu Y, Wang J, Pi J. Differential analysis of ovarian tissue between high and low-yielded laying hens in the late laying stage and the effect of LECT2 gene on follicular granulosa cells proliferation. Mol Biol Rep 2024; 51:240. [PMID: 38300380 DOI: 10.1007/s11033-024-09260-8] [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: 09/15/2023] [Accepted: 01/15/2024] [Indexed: 02/02/2024]
Abstract
The ovaries of high-yield laying hens exhibited signs of aging beyond 400 days of age, subsequently resulting in a decline in both egg production and egg quality. Oxidative stress, characterized by an increase in the production of reactive oxygen species (ROS), stands as one of the principal processes contributing to ovarian aging. Elevated ROS levels are implicated in the induction of apoptosis in granulosa cells (GCs), provoking mitochondrial impairment, and diminishing the capacity of the antioxidant defense system. This investigation stratified laying hens into two distinct groups, predicated upon their egg production levels: high-yield hens (HH) and low-yield hens (LL). The study focused on evaluating oxidative stress markers and identifying differentially expressed genes between these two groups. The findings revealed that the LL group exhibited follicular atresia, mitochondrial disruptions, and apoptotic occurrences in ovarian GCs. Notably, ROS levels, Malondialdehyde (MDA) concentrations, and 8-hydroxy-2'-deoxyguanosine (8-OHdG) concentrations in ovarian tissue and follicular GCs were substantially higher in the HH group. Furthermore, the RNA-sequencing results unveiled differential expression of the LECT2 gene between the HH and LL groups. Consequently, an overexpression vector for the LECT2 gene was successfully constructed and introduced into GCs. The quantitative polymerase chain reaction (QPCR) analysis exhibited significant downregulation (p < 0.01) of key apoptotic genes such as Caspase-3 and C-myc and significant upregulation (p < 0.01) of BCL2 following the overexpression of the LECT2 gene in GCs. In conclusion, oxidative stress emerges as a pivotal factor influencing the laying traits of both high and low-yield laying hens. The accumulation of reactive oxygen species (ROS) within the ovaries precipitates apoptosis in GCs, subsequently leading to follicular atresia and a reduction in egg production. Furthermore, we employed RNA sequencing technology to examine the ovarian matrix tissue in high and low laying hens during the late phase of egg laying. Our analysis revealed a substantial upregulation of the LECT2 gene in the ovarian matrix tissue of high laying hens. This observation implies that the LECT2 gene exerts a pivotal influence on driving the proliferation and differentiation of follicular GCs, thereby exerting a crucial regulatory role in follicular development.
Collapse
Affiliation(s)
- Lin Chen
- Animal Husbandry and Veterinary Research Institute, Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Hubei Academy of Agricultural Sciences, Wuhan, 430000, China
- College of Animal Science, Yangtze University, Jingzhou, 434000, China
| | - Xianqiang Li
- Animal Husbandry and Veterinary Research Institute, Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Hubei Academy of Agricultural Sciences, Wuhan, 430000, China
- College of Animal Science, Yangtze University, Jingzhou, 434000, China
| | - Yan Wu
- Animal Husbandry and Veterinary Research Institute, Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Hubei Academy of Agricultural Sciences, Wuhan, 430000, China.
| | - Jiaxiang Wang
- College of Animal Science, Yangtze University, Jingzhou, 434000, China
| | - Jinsong Pi
- Animal Husbandry and Veterinary Research Institute, Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Hubei Academy of Agricultural Sciences, Wuhan, 430000, China
| |
Collapse
|
28
|
Zhu P, Wu X, Ni L, Chen K, Dong Z, Du J, Kong F, Mao Y, Tao H, Chu M, Mao H, Yang H, Liu Q, Gan M, Geng D. Inhibition of PP2A ameliorates intervertebral disc degeneration by reducing annulus fibrosus cells apoptosis via p38/MAPK signal pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166888. [PMID: 37722489 DOI: 10.1016/j.bbadis.2023.166888] [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: 04/12/2023] [Revised: 07/05/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is considered one of the main reasons for low back pain (LBP). To date, the specific pathology of IVDD remains unclear. The annulus fibrosus (AF) is an important part of the intervertebral disc, and AF cell oxidative stress, apoptosis plays a vital role in disc degeneration. Protein phosphatase 2 A (PP2A), a serine/threonine phosphatase, has regulatory functions in various processes, including apoptosis and autophagy. However, thus far, the effect of PP2A on IVDD is not clear. METHODS AF cells derived from caudal intervertebral discs in SD rats were used to analyze the levels of oxidative stress, apoptosis and degeneration as well as PP2A expression. A PP2A agonist (FTY720), inhibitor (microcystin-LR) and siRNA (si-PPP2CA) were employed in IVDD induced by H2O2 to investigate the levels of apoptosis and degeneration. The p38/MAPK signal pathways were evaluated, and a p38 inhibitor (SB203580) and ERK inhibitor (U0126) were added for verification. Finally, FTY720 and microcystin-LR were administered to IVDD rats to assess the effects on levels of apoptosis and degeneration and the relief of IVDD. RESULTS The expression of PP2A was increased in rat AF cells after H2O2 intervention. The levels of apoptosis and degeneration were higher with upregulation of PP2A but were significantly reduced after inhibition of PP2A. The PP2A inhibitor relieved cell apoptosis and degeneration by downregulating the p38/MAPK pathway. In vivo, the knockdown of PP2A resulted in a more complete morphology of discs and less apoptotic and degenerative expression. CONCLUSIONS This study suggests that the downregulation of PP2 A could reduce AF cell apoptosis and degeneration via the p38/MAPK pathway. It also revealed that the inhibition of PP2 A is expected to be a therapeutic target for IVDD.
Collapse
Affiliation(s)
- Pengfei Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Xiexing Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Li Ni
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Kai Chen
- Department of Orthopedics, Hai'an People's Hospital, Hai'an 226600, Jiangsu, China
| | - Zhongchen Dong
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Jiacheng Du
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Fanchen Kong
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yubo Mao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Huaqiang Tao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Miao Chu
- Department of Orthopedics, Yixing People's Hospital, Yixing 214200, Jiangsu, China
| | - Haiqin Mao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Qinbai Liu
- Department of Orthopaedics, Lianshui People's Hospital of Kangda College Affiliated to Nanjing Medical University, Huai'an 223001, Jiangsu, China; Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| | - Minfeng Gan
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
29
|
Alzahrani SA, Bekhet GM, Ammar RB, Abdallah BM, Ali EM, Al-Ramadan SY, Althumairy D, Rajendran P. The Inhibitory Effect of Geraniol on CCL4-induced Hepatorenal Toxicity in Pregnant Mice through the PI3K/AKT Signaling Pathway. SAUDI JOURNAL OF MEDICINE & MEDICAL SCIENCES 2024; 12:17-26. [PMID: 38362098 PMCID: PMC10866391 DOI: 10.4103/sjmms.sjmms_225_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/21/2023] [Accepted: 10/08/2023] [Indexed: 02/17/2024]
Abstract
Background Hepatotoxicity caused by CCL4 is well known. Geraniol (GNL) has high antioxidant effect that can induces liver regeneration. However, the protective effect of GNL effect on CCL4-induced hepatorenal toxicity in pregnant mice has not yet been studied. Objective To investigate whether GNL could protect against oxidative stress induced by CCL4 via the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, which is regulated by phosphatidylinositol 3 kinase/protein kinase B (PI3K/AKT), and has been found to have protective effects on renal and hepatic tissues. Materials and Methods Forty-eight female albino mice weighing 25-30 g were randomly allocated to 4 groups: Group I served as a control; Group II received a toxicity-inducing single dose of 15 μL of CCL4 on the 4th day after mating; Group III received 40 mg/kg GNL + CCL4 (with GNL from the 1st day of assimilation to delivery); and Group IV received GNL alone from the 1st day of assimilation to the end of the delivery period. GNL was evaluated for its protective effects on hepatotoxicity in CCL4-treated pregnant mice. Litter size, weight, survival rate, and resorption were recorded. In addition, H & E staining was done for liver and kidney pathology as well as biochemical markers and oxidative markers malondialdehyde, superoxide dismutase, and catalase were analyzed. Results CCL4 significantly reduced survival rate and increased resorption after exposure. Alanine transaminase and aspartate aminotransferase concentrations in the serum, tissue MDA, blood urea nitrogen, and creatinine were increased after CCL4 exposure. GNL improved enzyme and antioxidant levels and prevented CCL4-induced hepatic injury in mice. Caspase-3 cleavage was decreased by GNL, which increased PI3K, phosphorylated AKT, Nrf2, and B-cell lymphoma 2. Conclusion GNL demonstrates a protective effect against CCl4-induced hepatorenal toxicity, mediated through the activation of the PI3K/AKT signaling pathway and the upregulation of Nrf2. These findings highlight the potential therapeutic implications of GNL in mitigating oxidative stress and inflammation in liver and kidney tissues.
Collapse
Affiliation(s)
- Sabah Ali Alzahrani
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Gamal M. Bekhet
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Zoology, Faculty of Science, Alexandria University Egypt, Alexandria, Egypt
| | - Rebai Ben Ammar
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
- Laboratory of Aromatic and Medicinal Plants, Center of Biotechnology of Borj Cedria, Hammam-Lif, Tunisia
| | - Basem M. Abdallah
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Enas Mohamed Ali
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Saeed Y. Al-Ramadan
- Department of Anatomy, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Duaa Althumairy
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Biochemistry, Centre of Molecular Medicine and Diagnostics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
30
|
Mohammed OA, Alghamdi M, Alfaifi J, Alamri MMS, Al-Shahrani AM, Alharthi MH, Alshahrani AM, Alhalafi AH, Adam MIE, Bahashwan E, Jarallah AlQahtani AA, BinAfif WF, Abdel-Reheim MA, Abdel Mageed SS, Doghish AS. The emerging role of miRNAs in myocardial infarction: From molecular signatures to therapeutic targets. Pathol Res Pract 2024; 253:155087. [PMID: 38183820 DOI: 10.1016/j.prp.2023.155087] [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: 12/20/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024]
Abstract
Globally, myocardial infarction (MI) and other cardiovascular illnesses have long been considered the top killers. Heart failure and mortality are the results of myocardial apoptosis, cardiomyocyte fibrosis, and cardiomyocyte hypertrophy, all of which are caused by MI. MicroRNAs (miRNAs) play a crucial regulatory function in the progression and advancement of heart disease following an MI. By consolidating the existing data on miRNAs, our aim is to gain a more comprehensive understanding of their role in the pathological progression of myocardial injury after MI and to identify potential crucial target pathways. Also included are the primary treatment modalities and their most recent developments. miRNAs have the ability to regulate both normal and pathological activity, including the key signaling pathways. As a result, they may exert medicinal benefits. This review presents a comprehensive analysis of the role of miRNAs in MI with a specific emphasis on their impact on the regeneration of cardiomyocytes and other forms of cell death, such as apoptosis, necrosis, and autophagy. Furthermore, the targets of pro- and anti-MI miRNAs are comparatively elucidated.
Collapse
Affiliation(s)
- Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Mushabab Alghamdi
- Department of Internal Medicine, Division of Rheumatology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Jaber Alfaifi
- Department of Child Health, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohannad Mohammad S Alamri
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Abdullah M Al-Shahrani
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Muffarah Hamid Alharthi
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Abdullah M Alshahrani
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Abdullah Hassan Alhalafi
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Masoud I E Adam
- Department of Medical Education and Internal Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Emad Bahashwan
- Department of Internal Medicine, Division of Dermatology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - AbdulElah Al Jarallah AlQahtani
- Department of Internal Medicine, Division of Dermatology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Waad Fuad BinAfif
- Department of Internal Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| |
Collapse
|
31
|
Chen RJ, Chen MC, Tsai BCK, Roy R, Chang YR, Wang TF, Kuo WW, Kuo CH, Yao CH, Li CC, Huang CY. Ligustrazine improves the compensative effect of Akt survival signaling to protect liver Kupffer cells in trauma-hemorrhagic shock rats. Chem Biol Drug Des 2023; 102:1399-1408. [PMID: 37612133 DOI: 10.1111/cbdd.14327] [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: 04/11/2023] [Revised: 07/16/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023]
Abstract
Trauma-hemorrhagic shock (THS) is a medical emergency that is encountered by physicians in the emergency department. Chuan Xiong is a traditional Chinese medicine and ligustrazine is a natural compound from it. Ligustrazine improves coronary blood flow and reduces cardiac ischemia in animals through Ca2+ and ATP-dependent vascular relaxation. It also decreases the platelets' bioactivity and reduces reactive oxygen species formation. We hypothesized that ligustrazine could protect liver by decreasing the inflammation response, protein production, and apoptosis in THS rats. Ligustrazine at doses of 100 and 1000 μg/mL was administrated in Kupffer cells isolated from THS rats. The protein expressions were detected via western blot. The THS showed increased inflammation response proteins, mitochondria-dependent apoptosis proteins, and had a compensation effect on the Akt pathway. After ligustrazine treatment, the hemorrhagic shock Kupffer cells decreased inflammatory response and mitochondria-dependent apoptosis and promoted a more compensative effect of the Akt pathway. It suggests ligustrazine reduces inflammation response and mitochondria-dependent apoptosis induced by THS in liver Kupffer cells and promotes more survival effects by elevating the Akt pathway. These findings demonstrate the beneficial effects of ligustrazine against THS-induced hepatic injury, and ligustrazine could be a potential medication to treat the liver injury caused by THS.
Collapse
Affiliation(s)
- Ray-Jade Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Bruce Chi-Kang Tsai
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Rakesh Roy
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yi-Ru Chang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tso-Fu Wang
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - Chun-Hsu Yao
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Biomaterials Translational Research Center, China Medical University Hospital, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Chi-Cheng Li
- School of Medicine, Tzu Chi University, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Center of Stem Cell & Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
| |
Collapse
|
32
|
Chong SJF, Zhu F, Dashevsky O, Mizuno R, Lai JX, Hackett L, Ryan CE, Collins MC, Iorgulescu JB, Guièze R, Penailillo J, Carrasco R, Hwang YC, Muñoz DP, Bouhaddou M, Lim YC, Wu CJ, Allan JN, Furman RR, Goh BC, Pervaiz S, Coppé JP, Mitsiades CS, Davids MS. Hyperphosphorylation of BCL-2 family proteins underlies functional resistance to venetoclax in lymphoid malignancies. J Clin Invest 2023; 133:e170169. [PMID: 37751299 PMCID: PMC10645378 DOI: 10.1172/jci170169] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023] Open
Abstract
The B cell leukemia/lymphoma 2 (BCL-2) inhibitor venetoclax is effective in chronic lymphocytic leukemia (CLL); however, resistance may develop over time. Other lymphoid malignancies such as diffuse large B cell lymphoma (DLBCL) are frequently intrinsically resistant to venetoclax. Although genomic resistance mechanisms such as BCL2 mutations have been described, this probably only explains a subset of resistant cases. Using 2 complementary functional precision medicine techniques - BH3 profiling and high-throughput kinase activity mapping - we found that hyperphosphorylation of BCL-2 family proteins, including antiapoptotic myeloid leukemia 1 (MCL-1) and BCL-2 and proapoptotic BCL-2 agonist of cell death (BAD) and BCL-2 associated X, apoptosis regulator (BAX), underlies functional mechanisms of both intrinsic and acquired resistance to venetoclax in CLL and DLBCL. Additionally, we provide evidence that antiapoptotic BCL-2 family protein phosphorylation altered the apoptotic protein interactome, thereby changing the profile of functional dependence on these prosurvival proteins. Targeting BCL-2 family protein phosphorylation with phosphatase-activating drugs rewired these dependencies, thus restoring sensitivity to venetoclax in a panel of venetoclax-resistant lymphoid cell lines, a resistant mouse model, and in paired patient samples before venetoclax treatment and at the time of progression.
Collapse
MESH Headings
- Mice
- Animals
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Drug Resistance, Neoplasm/genetics
- Proto-Oncogene Proteins c-bcl-2/genetics
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- bcl-X Protein/genetics
- Apoptosis Regulatory Proteins
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Cell Line, Tumor
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
Collapse
Affiliation(s)
- Stephen Jun Fei Chong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Fen Zhu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Olga Dashevsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Rin Mizuno
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jolin X.H. Lai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Liam Hackett
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Christine E. Ryan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Mary C. Collins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - J. Bryan Iorgulescu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Romain Guièze
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Johany Penailillo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Ruben Carrasco
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Yeonjoo C. Hwang
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Denise P. Muñoz
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, California, USA
| | - Yaw Chyn Lim
- Cancer Science Institute, National University of Singapore, Singapore
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - John N. Allan
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Richard R. Furman
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Boon Cher Goh
- Cancer Science Institute, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jean-Philippe Coppé
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Constantine S. Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew S. Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Al-Karmalawy AA, Rashed M, Sharaky M, Abulkhair HS, Hammouda MM, Tawfik HO, Shaldam MA. Novel fused imidazotriazines acting as promising top. II inhibitors and apoptotic inducers with greater selectivity against head and neck tumors: Design, synthesis, and biological assessments. Eur J Med Chem 2023; 259:115661. [PMID: 37482023 DOI: 10.1016/j.ejmech.2023.115661] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/08/2023] [Accepted: 07/16/2023] [Indexed: 07/25/2023]
Abstract
Although the great effectiveness of doxorubicin (Dox) in the treatment of many types of tumors, it showed limited effectiveness against the head and neck squamous cell carcinoma (HNSCC) subtype which is attributed to its reported multiple drug resistance (MDR). In the current study, we considered the essential pharmacophoric features of Dox as an effective Top. II inhibitor and sought to develop a novel set of imidazo[1,2-a] [1,3,5]triazin-2-amines (2a-2p) as a suggested anticancer option that could intercalate the DNA base pairs. We evaluated the % inhibition of the newly synthesized compounds on thirteen cancer cell lines and the analysis of structure-activity relationships revealed that the human head and neck cancer cell line (HNO97) was the most sensitive to their growth inhibition effect. Then, the IC50 values were recorded against the most sensitive cancer cell lines (HNO97, MDA-MB-231, and HEPG2), and compared to the normal cell line OEC (human oral epithelial cells). Compounds 2f and 2g showed very strong activities against HNO97 with IC50 values of (4 ± 1 and 3 ± 1.5 μg/mL), respectively, compared to that of Dox (9 ± 1.6 μg/mL). Next, a quantitative determination of human DNA Top. II concentrations in the most sensitive cell line (HNO97) were recorded for the most active anticancer derivatives. Again, compound 2f showed a superior Top. II inhibition with 87.86% compared to that of Dox (86.44%), while compound 2g achieved an inhibition of 81.37% which was close to the effect of Dox. To further investigate their effects on cell cycle progression and apoptosis induction in HNO97 cells, both 2f and 2g were selected for analysis. Both candidates arrested cell cycle progression at both the S and G2-M phases, as well as increased the early and late apoptosis phase ratios. Besides, both 2f and 2g were subjected to protein expression analysis of apoptosis-related genes (p53, BAX, IL-6, and BCL2). Moreover, the antioxidant effect of 2f and 2g was evaluated by measuring GSH, MDA, and NO markers in HNO97 cells. Furthermore, molecular docking for the newly designed tricyclic derivatives against both the Top. II and DNA double helix was carried out.
Collapse
Affiliation(s)
- Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, 12566, Egypt.
| | - Mahmoud Rashed
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, 11884, Cairo, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University - Egypt, International Coastal Road, New Damietta, 34518, Egypt
| | - Mohamed M Hammouda
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia; Department of Chemistry, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| |
Collapse
|
34
|
Mendez-Callejas G, Piñeros-Avila M, Yosa-Reyes J, Pestana-Nobles R, Torrenegra R, Camargo-Ubate MF, Bello-Castro AE, Celis CA. A Novel Tri-Hydroxy-Methylated Chalcone Isolated from Chromolaena tacotana with Anti-Cancer Potential Targeting Pro-Survival Proteins. Int J Mol Sci 2023; 24:15185. [PMID: 37894866 PMCID: PMC10607159 DOI: 10.3390/ijms242015185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Chromolaena tacotana (Klatt) R. M. King and H. Rob (Ch. tacotana) contains bioactive flavonoids that may have antioxidant and/or anti-cancer properties. This study investigated the potential anti-cancer properties of a newly identified chalcone isolated from the inflorescences of the plant Chromolaena tacotana (Klatt) R. M. King and H. Rob (Ch. tacotana). The chalcone structure was determined using HPLC/MS (QTOF), UV, and NMR spectroscopy. The compound cytotoxicity and selectivity were evaluated on prostate, cervical, and breast cancer cell lines using the MTT assay. Apoptosis and autophagy induction were assessed through flow cytometry by detecting annexin V/7-AAD, active Casp3/7, and LC3B proteins. These results were supported by Western blot analysis. Mitochondrial effects on membrane potential, as well as levels of pro- and anti-apoptotic proteins were analyzed using flow cytometry, fluorescent microscopy, and Western blot analysis specifically on a triple-negative breast cancer (TNBC) cell line. Furthermore, molecular docking (MD) and molecular dynamics (MD) simulations were performed to evaluate the interaction between the compounds and pro-survival proteins. The compound identified as 2',3,4-trihydroxy-4',6'-dimethoxy chalcone inhibited the cancer cell line proliferation and induced apoptosis and autophagy. MDA-MB-231, a TNBC cell line, exhibited the highest sensitivity to the compound with good selectivity. This activity was associated with the regulation of mitochondrial membrane potential, activation of the pro-apoptotic proteins, and reduction of anti-apoptotic proteins, thereby triggering the intrinsic apoptotic pathway. The chalcone consistently interacted with anti-apoptotic proteins, particularly the Bcl-2 protein, throughout the simulation period. However, there was a noticeable conformational shift observed with the negative autophagy regulator mTOR protein. Future studies should focus on the molecular mechanisms underlying the anti-cancer potential of the new chalcone and other flavonoids from Ch. tacotana, particularly against predominant cancer cell types.
Collapse
Affiliation(s)
- Gina Mendez-Callejas
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada (GIBGA), Laboratorio de Biología Celular y Molecular, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 # 55-37, Bogotá 111166, Colombia;
| | - Marco Piñeros-Avila
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada (GIBGA), Laboratorio de Biología Celular y Molecular, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 # 55-37, Bogotá 111166, Colombia;
| | - Juvenal Yosa-Reyes
- Grupo de Investigación en Ciencias Exactas, Física y Naturales Aplicadas, Facultad de Ciencias Básicas y Biomédicas, Laboratorio de Simulación Molecular y Bioinformática, Universidad Simón Bolívar, Carrera 59 # 59-65, Barranquilla 080002, Colombia; (J.Y.-R.)
| | - Roberto Pestana-Nobles
- Grupo de Investigación en Ciencias Exactas, Física y Naturales Aplicadas, Facultad de Ciencias Básicas y Biomédicas, Laboratorio de Simulación Molecular y Bioinformática, Universidad Simón Bolívar, Carrera 59 # 59-65, Barranquilla 080002, Colombia; (J.Y.-R.)
| | - Ruben Torrenegra
- Grupo de Investigación en Productos Naturales de la U.D.C.A. (PRONAUDCA), Laboratorio de Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 # 55-37, Bogotá 111166, Colombia
| | - María F. Camargo-Ubate
- Grupo de Investigación en Productos Naturales de la U.D.C.A. (PRONAUDCA), Laboratorio de Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 # 55-37, Bogotá 111166, Colombia
| | - Andrea E. Bello-Castro
- Grupo de Investigación en Productos Naturales de la U.D.C.A. (PRONAUDCA), Laboratorio de Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 # 55-37, Bogotá 111166, Colombia
| | - Crispin A. Celis
- Grupo de Investigación en Fitoquímica (GIFUJ), Departamento de Química, Facultad de Ciencias, Pontificia Universidad Javeriana, Cra. 7 # 40-62, Bogotá 1115511, Colombia
| |
Collapse
|
35
|
Peroni E, Randi ML, Rosato A, Cagnin S. Acute myeloid leukemia: from NGS, through scRNA-seq, to CAR-T. dissect cancer heterogeneity and tailor the treatment. J Exp Clin Cancer Res 2023; 42:259. [PMID: 37803464 PMCID: PMC10557350 DOI: 10.1186/s13046-023-02841-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a malignant blood cancer with marked cellular heterogeneity due to altered maturation and differentiation of myeloid blasts, the possible causes of which are transcriptional or epigenetic alterations, impaired apoptosis, and excessive cell proliferation. This neoplasm has a high rate of resistance to anticancer therapies and thus a high risk of relapse and mortality because of both the biological diversity of the patient and intratumoral heterogeneity due to the acquisition of new somatic changes. For more than 40 years, the old gold standard "one size fits all" treatment approach included intensive chemotherapy treatment with anthracyclines and cytarabine.The manuscript first traces the evolution of the understanding of the pathology from the 1970s to the present. The enormous strides made in its categorization prove to be crucial for risk stratification, enabling an increasingly personalized diagnosis and treatment approach.Subsequently, we highlight how, over the past 15 years, technological advances enabling single cell RNA sequencing and T-cell modification based on the genomic tools are affecting the classification and treatment of AML. At the dawn of the new millennium, the advent of high-throughput next-generation sequencing technologies has enabled the profiling of patients evidencing different facets of the same disease, stratifying risk, and identifying new possible therapeutic targets that have subsequently been validated. Currently, the possibility of investigating tumor heterogeneity at the single cell level, profiling the tumor at the time of diagnosis or after treatments exist. This would allow the identification of underrepresented cellular subclones or clones resistant to therapeutic approaches and thus responsible for post-treatment relapse that would otherwise be difficult to detect with bulk investigations on the tumor biopsy. Single-cell investigation will then allow even greater personalization of therapy to the genetic and transcriptional profile of the tumor, saving valuable time and dangerous side effects. The era of personalized medicine will take a huge step forward through the disclosure of each individual piece of the complex puzzle that is cancer pathology, to implement a "tailored" therapeutic approach based also on engineered CAR-T cells.
Collapse
Affiliation(s)
- Edoardo Peroni
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Padova, 35128, Italy.
| | - Maria Luigia Randi
- First Medical Clinic, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Padova, 35128, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Stefano Cagnin
- Department of Biology, University of Padova, Padova, 35131, Italy
- CIR-Myo Myology Center, University of Padova, Padova, 35131, Italy
| |
Collapse
|
36
|
Wang Z, Guo X, Lian J, Ji Y, Li K. Prognostic value of amino acid metabolism-related gene expression in invasive breast carcinoma. J Cancer Res Clin Oncol 2023; 149:11117-11133. [PMID: 37340191 DOI: 10.1007/s00432-023-04985-8] [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: 03/22/2023] [Accepted: 06/13/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND An increasing number of studies indicated that metabolic reprogramming of amino acid metabolism may either promote or inhibit tumor progression. The purpose of this study was to investigate the ability of a gene risk signature associated with amino acid metabolism to predict the prognosis and immune characteristics of invasive breast carcinoma. METHODS LASSO Cox regression analysis was performed to construct and validate the prognostic risk signature based on the expression of 9 amino acid metabolism-related genes. The predictive value of the signature, immune characteristics, and chemotherapeutic drugs was also predicted. Finally, 9 significant genes were examined in MDA-MB-231 and MCF-7 cells, and the predicted chemotherapeutic drugs were also verified. RESULTS The prognosis of the low-risk group was better than that of the high-risk group. The areas under the curve (AUCs) at 1, 2, and 3 years were 0.852, 0.790, and 0.736, respectively. In addition, the GSEA results for KEGG and GO revealed that samples with a high-risk score exhibited a variety of highly malignant manifestations. The high-risk group was characterized by an increased number of M2 macrophages, a high level of tumor purity, low levels of APC co-stimulation, cytolytic activity, HLA, para-inflammation, and type I IFN response. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) confirmed that MDA-MB-231 and MCF-7 cells express 9 amino acid metabolism-related genes differently. In addition, cell experiments were conducted to examine the effect of cephaeline-induced on cell viability, migration ability, and protein expression of the PI3K/AKT signaling pathway and HIF-1α. CONCLUSION We established a risk signature based on 9 amino acid metabolism-related genes for invasive breast carcinoma. Further analyses revealed that this risk signature is superior to other clinical indexes in survival prediction and that the subgroups identified by the risk signature exhibit distinct immune characteristics. Cephaeline was determined to be a superior option for patients in high-risk groups.
Collapse
Affiliation(s)
- Zilin Wang
- Department of Radiology, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, No. 650 New Songjiang Road, Shanghai, 200080, People's Republic of China
| | - Xinyu Guo
- Department of Radiology, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, No. 650 New Songjiang Road, Shanghai, 200080, People's Republic of China
| | - Jingge Lian
- Department of Radiology, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, No. 650 New Songjiang Road, Shanghai, 200080, People's Republic of China
| | - Ying Ji
- Department of Radiology, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, No. 650 New Songjiang Road, Shanghai, 200080, People's Republic of China
| | - Kangan Li
- Department of Radiology, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, No. 650 New Songjiang Road, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
37
|
Peris I, Romero-Murillo S, Vicente C, Narla G, Odero MD. Regulation and role of the PP2A-B56 holoenzyme family in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188953. [PMID: 37437699 DOI: 10.1016/j.bbcan.2023.188953] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
Protein phosphatase 2A (PP2A) inactivation is common in cancer, leading to sustained activation of pro-survival and growth-promoting pathways. PP2A consists of a scaffolding A-subunit, a catalytic C-subunit, and a regulatory B-subunit. The functional complexity of PP2A holoenzymes arises mainly through the vast repertoire of regulatory B-subunits, which determine both their substrate specificity and their subcellular localization. Therefore, a major challenge for developing more effective therapeutic strategies for cancer is to identify the specific PP2A complexes to be targeted. Of note, the development of small molecules specifically directed at PP2A-B56α has opened new therapeutic avenues in both solid and hematological tumors. Here, we focus on the B56/PR61 family of PP2A regulatory subunits, which have a central role in directing PP2A tumor suppressor activity. We provide an overview of the mechanisms controlling the formation and regulation of these complexes, the pathways they control, and the mechanisms underlying their deregulation in cancer.
Collapse
Affiliation(s)
- Irene Peris
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain; Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| | - Silvia Romero-Murillo
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain; Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
| | - Carmen Vicente
- Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, MI, USA
| | - Maria D Odero
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain; Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
38
|
Harris DD, Sabe SA, Sabra M, Xu CM, Malhotra A, Broadwin M, Banerjee D, Abid MR, Sellke FW. Intramyocardial injection of hypoxia-conditioned extracellular vesicles modulates apoptotic signaling in chronically ischemic myocardium. JTCVS OPEN 2023; 15:220-228. [PMID: 37808040 PMCID: PMC10556811 DOI: 10.1016/j.xjon.2023.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/28/2023] [Accepted: 05/18/2023] [Indexed: 10/10/2023]
Abstract
Objective Limited treatments exist for nonoperative chronic coronary artery disease. Previously, our laboratory has investigated extracellular vesicle (EV) therapy as a potential treatment for chronic coronary artery disease using a swine model and demonstrated improved cardiac function in swine treated with intramyocardial EV injection. Here, we seek to investigate the potential cardiac benefits of EVs by using hypoxia-conditioned EVs (HEV). Specifically, this study aims to investigate the effect of HEV on apoptosis in chronically ischemic myocardium in swine. Methods Fourteen Yorkshire swine underwent placement of an ameroid constrictor on the left circumflex artery. Two weeks later, swine underwent redo left thoracotomy with injection of either saline (control, n = 7) or HEVs (n = 7). After 5 weeks, swine were euthanized for tissue collection. Terminal deoxynucleotidyl transferase dUTP nick end labeling was used to quantify apoptosis. Immunoblotting was used for protein quantification. Results Terminal deoxynucleotidyl transferase dUTP nick end labeling staining showed a decrease in apoptosis in the HEV group compared with the control (P = .049). The HEV group exhibited a significant increase in the anti-apoptotic signaling molecule phospho-BAD (P = .005), a significant decrease in B-cell lymphoma 2 (P = .006) and an increase in the phospho-B-cell lymphoma to B-cell lymphoma 2 ratio (P < .001). Furthermore, the HEV group exhibited increased levels of prosurvival signaling markers including phosphoinositide 3-kinase, phosphor-extracellular signal-regulated kinase 1/2, phospho-forkhead box protein O1, and phospho-protein kinase B to protein kinase B ratio (all P < .05). Conclusions In chronic myocardial ischemia, treatment with HEV results in a decrease in overall apoptosis, possibly through the activation of both pro-survival and anti-apoptotic signaling pathways.
Collapse
Affiliation(s)
- Dwight D. Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Sharif A. Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Mohamed Sabra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Cynthia M. Xu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Akshay Malhotra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Debolina Banerjee
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| |
Collapse
|
39
|
Elsakka EGE, Abulsoud AI, El-Mahdy HA, Ismail A, Elballal MS, Mageed SSA, Khidr EG, Mohammed OA, Sarhan OM, Elkhawaga SY, El-Husseiny AA, Abdelmaksoud NM, El-Demerdash AA, Shahin RK, Midan HM, Elrebehy MA, Doghish AA, Doghish AS. miRNAs orchestration of cardiovascular diseases - Particular emphasis on diagnosis, and progression. Pathol Res Pract 2023; 248:154613. [PMID: 37327567 DOI: 10.1016/j.prp.2023.154613] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/18/2023]
Abstract
MicroRNAs (miRNAs; miRs) are small non-coding ribonucleic acids sequences vital in regulating gene expression. They are significant in many biological and pathological processes and are even detectable in various body fluids such as serum, plasma, and urine. Research has demonstrated that the irregularity of miRNA in multiplying cardiac cells is linked to developmental deformities in the heart's structure. It has also shown that miRNAs are crucial in diagnosing and progressing several cardiovascular diseases (CVDs). The review covers the function of miRNAs in the pathophysiology of CVD. Additionally, the review provides an overview of the potential role of miRNAs as disease-specific diagnostic and prognostic biomarkers for human CVD, as well as their biological implications in CVD.
Collapse
Affiliation(s)
- Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Emad Gamil Khidr
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Osama A Mohammed
- Department of Clinical Pharmacology, Faculty of Medicine, Bisha University, Bisha 61922, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Omnia M Sarhan
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Samy Y Elkhawaga
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, 11829 Cairo, Egypt
| | | | - Aya A El-Demerdash
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Reem K Shahin
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Heba M Midan
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ayman A Doghish
- Department of Cardiovascular & Thoracic Surgery, Ain-Shams University Hospital, Faculty of Medicine, Cairo, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| |
Collapse
|
40
|
Griswold-Prenner I, Kashyap AK, Mazhar S, Hall ZW, Fazelinia H, Ischiropoulos H. Unveiling the human nitroproteome: Protein tyrosine nitration in cell signaling and cancer. J Biol Chem 2023; 299:105038. [PMID: 37442231 PMCID: PMC10413360 DOI: 10.1016/j.jbc.2023.105038] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/28/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023] Open
Abstract
Covalent amino acid modification significantly expands protein functional capability in regulating biological processes. Tyrosine residues can undergo phosphorylation, sulfation, adenylation, halogenation, and nitration. These posttranslational modifications (PTMs) result from the actions of specific enzymes: tyrosine kinases, tyrosyl-protein sulfotransferase(s), adenylate transferase(s), oxidoreductases, peroxidases, and metal-heme containing proteins. Whereas phosphorylation, sulfation, and adenylation modify the hydroxyl group of tyrosine, tyrosine halogenation and nitration target the adjacent carbon residues. Because aberrant tyrosine nitration has been associated with human disorders and with animal models of disease, we have created an updated and curated database of 908 human nitrated proteins. We have also analyzed this new resource to provide insight into the role of tyrosine nitration in cancer biology, an area that has not previously been considered in detail. Unexpectedly, we have found that 879 of the 1971 known sites of tyrosine nitration are also sites of phosphorylation suggesting an extensive role for nitration in cell signaling. Overall, the review offers several forward-looking opportunities for future research and new perspectives for understanding the role of tyrosine nitration in cancer biology.
Collapse
Affiliation(s)
| | | | | | - Zach W Hall
- Nitrase Therapeutics, Brisbane, California, USA
| | - Hossein Fazelinia
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
41
|
Ma G, Zhang J, Wang Z, Yu Q, Han L. Effects of muscle-specific oxidative stress on protein phosphorylation and its relationship with mitochondrial dysfunction, muscle oxidation, and apoptosis. Food Chem 2023; 427:136737. [PMID: 37390736 DOI: 10.1016/j.foodchem.2023.136737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/23/2023] [Accepted: 06/24/2023] [Indexed: 07/02/2023]
Abstract
This study aimed to investigate the effects of muscle-specific oxidative stress on phosphorylation and its relationship with mitochondrial dysfunction, muscle oxidation, and apoptosis of porcine PM (psoas major) and LL (longissimus lumborum) during the first 24 h postmortem. The global phosphorylation level decreased and the mitochondrial dysfunction, oxidation level, and apoptosis increased significantly at 12 h postmortem compared with 2 h postmortem, suggesting that lower phosphorylation level was related to more mitochondrial dysfunction and apoptosis during the early postmortem, regardless of muscle type. PM exhibited a higher global phosphorylation level but showed greater mitochondrial dysfunction, oxidation level, and apoptosis than LL, regardless of aging time. The increased mitochondrial dysfunction and oxidative stress accelerated apoptosis, but their relationship with phosphorylation was different in various muscle types at different aging times. These findings provide insight regarding the roles of coordinated regulation of phosphorylation and apoptosis in development of quality of different muscles.
Collapse
Affiliation(s)
- Guoyuan Ma
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China.
| | - Jiaying Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Zhuo Wang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Qunli Yu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Ling Han
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
42
|
Al-Alem U, Rauscher GH, Alem QA, Kajdacsy-Balla A, Mahmoud AM. Prognostic Value of SGK1 and Bcl-2 in Invasive Breast Cancer. Cancers (Basel) 2023; 15:3151. [PMID: 37370761 DOI: 10.3390/cancers15123151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
It is crucial to understand molecular alterations in breast cancer and how they relate to clinicopathologic factors. We have previously shown that the glucocorticoid receptor (GCR) protein expression was reduced in invasive breast carcinoma compared to normal breast tissue. Glucocorticoids, signaling through the GCR, regulate several cellular processes via downstream targets such as serum/glucocorticoid-regulated kinase 1 (SGK1) and B-cell lymphoma 2 (Bcl-2). We measured the expression of SGK1 and Bcl-2, in respective breast cancer tissue arrays, from a multiracial cohort of breast cancer patients. Higher cytoplasmic SGK1 staining was stronger in breast cancer tissue compared to normal tissue, especially in hormone receptor-negative cases. Conversely, the expression of cytoplasmic Bcl-2 was reduced in breast cancer compared to normal tissue, especially in hormone receptor-negative cases. Bcl-2 staining was associated with the self-reported racial/ethnic category, an earlier clinical stage, a lower histological grade, and a higher survival rate. Bcl-2 expression was associated with longer survival in models adjusted for age and race (HR = 0.32, 95% CI: 0.15, 0.65), and Bcl-2 expression remained strongly positively associated with protection from breast cancer death, with additional adjustments for ER/PR status (HR = 0.41, 95% CI: 0.2, 0.85). SGK1 and Bcl-2 may play biological roles in breast cancer development and/or progression.
Collapse
Affiliation(s)
- Umaima Al-Alem
- Division of Epidemiology and Biostatistics, School of Public Health, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Garth H Rauscher
- Division of Epidemiology and Biostatistics, School of Public Health, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Qais Al Alem
- Division of Epidemiology and Biostatistics, School of Public Health, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Andre Kajdacsy-Balla
- Department of Pathology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Abeer M Mahmoud
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, The University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
43
|
Chernyuk D, Callens M, Polozova M, Gordeev A, Chigriai M, Rakovskaya A, Ilina A, Pchitskaya E, Van den Haute C, Vervliet T, Bultynck G, Bezprozvanny I. Neuroprotective properties of anti-apoptotic BCL-2 proteins in 5xFAD mouse model of Alzheimer's disease. IBRO Neurosci Rep 2023; 14:273-283. [PMID: 36926591 PMCID: PMC10011438 DOI: 10.1016/j.ibneur.2023.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/31/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. An early feature of the AD pathology is the dysregulation of intracellular Ca2+ signaling in neurons. In particular, increased Ca2+ release from endoplasmic reticulum-located Ca2+ channels, including inositol-1,4,5-trisphosphate type 1 receptors (IP3R1) and ryanodine receptors type 2 (RyR2), have been extensively reported. Known for its anti-apoptotic properties, Bcl-2 also has the ability to bind to and inhibit the Ca2+-flux properties of IP3Rs and RyRs. In this study, the hypothesis that the expression of Bcl-2 proteins can normalize dysregulated Ca2+ signaling in a mouse model of AD (5xFAD) and thereby prevent or slow the progression of AD was examined. Therefore, stereotactic injections of adeno-associated viral vectors expressing Bcl-2 proteins were performed in the CA1 region of the 5xFAD mouse hippocampus. In order to assess the importance of the association with IP3R1, the Bcl-2K17D mutant was also included in these experiments. This K17D mutation has been previously shown to decrease the association of Bcl-2 with IP3R1, thereby impairing its ability to inhibit IP3R1 while not affecting Bcl-2's ability to inhibit RyRs. Here, we demonstrate that Bcl-2 protein expression leads to synaptoprotective and amyloid-protective effects in the 5xFAD animal model. Several of these neuroprotective features are also observed by Bcl-2K17D protein expression, suggesting that these effects are not associated with Bcl-2-mediated inhibition of IP3R1. Potential mechanisms for this Bcl-2 synaptoprotective action may be related to its ability to inhibit RyR2 activity as Bcl-2 and Bcl-2K17D are equally potent in inhibiting RyR2-mediated Ca2+ fluxes. This work indicates that Bcl-2-based strategies hold neuroprotective potential in AD models, though the underlying mechanisms requires further investigation.
Collapse
Affiliation(s)
- D Chernyuk
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - M Callens
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - M Polozova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - A Gordeev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - M Chigriai
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - A Rakovskaya
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - A Ilina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - E Pchitskaya
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - C Van den Haute
- KU Leuven, Research Group for Neurobiology and Gene Therapy, Department of Neurosciences, Campus Gasthuisberg O/N-5 box 1023, Herestraat 49, BE-3000 Leuven, Belgium.,Leuven Viral Vector Core, BE-3000 Leuven, Belgium
| | - T Vervliet
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - G Bultynck
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - I Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia.,Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, USA
| |
Collapse
|
44
|
Sani F, Sani M, Moayedfard Z, Darayee M, Tayebi L, Azarpira N. Potential advantages of genetically modified mesenchymal stem cells in the treatment of acute and chronic liver diseases. Stem Cell Res Ther 2023; 14:138. [PMID: 37226279 DOI: 10.1186/s13287-023-03364-x] [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: 01/14/2023] [Accepted: 05/04/2023] [Indexed: 05/26/2023] Open
Abstract
Liver damage caused by toxicity can lead to various severe conditions, such as acute liver failure (ALF), fibrogenesis, and cirrhosis. Among these, liver cirrhosis (LC) is recognized as the leading cause of liver-related deaths globally. Unfortunately, patients with progressive cirrhosis are often on a waiting list, with limited donor organs, postoperative complications, immune system side effects, and high financial costs being some of the factors restricting transplantation. Although the liver has some capacity for self-renewal due to the presence of stem cells, it is usually insufficient to prevent the progression of LC and ALF. One potential therapeutic approach to improving liver function is the transplantation of gene-engineered stem cells. Several types of mesenchymal stem cells from various sources have been suggested for stem cell therapy for liver disease. Genetic engineering is an effective strategy that enhances the regenerative potential of stem cells by releasing growth factors and cytokines. In this review, we primarily focus on the genetic engineering of stem cells to improve their ability to treat damaged liver function. We also recommend further research into accurate treatment methods that involve safe gene modification and long-term follow-up of patients to increase the effectiveness and reliability of these therapeutic strategies.
Collapse
Affiliation(s)
- Farnaz Sani
- Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahsa Sani
- Department of Tissue Engineering and Cell Therapy, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Moayedfard
- Department of Tissue Engineering and Cell Therapy, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Darayee
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Khalili Street, P.O. Box: 7193711351, Shiraz, Iran.
| |
Collapse
|
45
|
Li H, Zhang Y, Hao Y, Xu P, Wang X, Zhu B, Lu C, Xu K. Proanthocyanidins Inhibit Osteoblast Apoptosis via the PI3K/AKT/Bcl-xL Pathway in the Treatment of Steroid-Induced Osteonecrosis of the Femoral Head in Rats. Nutrients 2023; 15:nu15081936. [PMID: 37111155 PMCID: PMC10140830 DOI: 10.3390/nu15081936] [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: 03/01/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Steroid-induced osteonecrosis of the femoral head (SONFH) is a common clinical disease caused by massive or prolonged use of steroids. Its pathogenesis is unclear, but its incidence is increasing annually. It is characterized by an insidious and rapid onset, and high disability rate, causing a great burden on patients' daily life. Therefore, clarifying its pathogenesis and providing early and effective treatment for steroid osteonecrosis is important. METHODS In vivo, we used methylprednisolone (MPS) to construct a SONFH rat model and employed Mirco-ct, Hematoxylin and eosin (H&E) staining, and TdT-mediated dUTP nick end labeling (TUNEL) staining analysis to evaluate the therapeutic effects of proanthocyanidins (PACs). Network pharmacology analysis was conducted to mine targets associated with femoral head necrosis, and PACs analyzed possible molecular mechanisms. In vitro, PACs were added at different doses after treatment of cells with dexamethasone (DEX), and human osteoblast-like sarcoma(MG-63) cell apoptosis was determined by Annexin V-FITC-PI. The mechanisms by which PACs regulate bone metabolism via the Phosphoinositide 3-kinase(PI3K)/protein kinase B(AKT)/Recombinant Human B-Cell Leukemia/Lymphoma 2 XL(Bcl-xL) axis were explored by Western blotting. RESULT In vivo studies showed that PACs prevented SONFH in rat model. The PI3K/AKT/Bcl-xL signaling pathway was selected by network pharmacology approach; in vitro studies showed that proanthocyanidin-activated AKT and Bcl-xL inhibited osteoblast apoptosis. CONCLUSIONS PACs can inhibit excessive osteoblast apoptosis in SONFH via the PI3K/AKT/Bcl-xL signaling axis and have potential therapeutic effects.
Collapse
Affiliation(s)
- Hui Li
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an 710054, China
- Department of Traditional Chinese and Western Medicine, First Clinical School of Shaanxi University of Traditional Chinese Medicine, Xianyang 712046, China
| | - Yufei Zhang
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an 710054, China
- Department of Traditional Chinese and Western Medicine, First Clinical School of Shaanxi University of Traditional Chinese Medicine, Xianyang 712046, China
| | - Yangquan Hao
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an 710054, China
| | - Peng Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an 710054, China
| | - Xingyu Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Bin Zhu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing 100000, China
| | - Chao Lu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an 710054, China
| | - Ke Xu
- Department of Traditional Chinese and Western Medicine, First Clinical School of Shaanxi University of Traditional Chinese Medicine, Xianyang 712046, China
| |
Collapse
|
46
|
Kim J, Ji S, Lee JY, Lorquin J, Orlikova-Boyer B, Cerella C, Mazumder A, Muller F, Dicato M, Detournay O, Diederich M. Marine Polyether Phycotoxin Palytoxin Induces Apoptotic Cell Death via Mcl-1 and Bcl-2 Downregulation. Mar Drugs 2023; 21:md21040233. [PMID: 37103372 PMCID: PMC10143546 DOI: 10.3390/md21040233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/28/2023] Open
Abstract
Palytoxin is considered one of the most potent biotoxins. As palytoxin-induced cancer cell death mechanisms remain to be elucidated, we investigated this effect on various leukemia and solid tumor cell lines at low picomolar concentrations. As palytoxin did not affect the viability of peripheral blood mononuclear cells (PBMC) from healthy donors and did not create systemic toxicity in zebrafish, we confirmed excellent differential toxicity. Cell death was characterized by a multi-parametric approach involving the detection of nuclear condensation and caspase activation assays. zVAD-sensitive apoptotic cell death was concomitant with a dose-dependent downregulation of antiapoptotic Bcl-2 family proteins Mcl-1 and Bcl-xL. Proteasome inhibitor MG-132 prevented the proteolysis of Mcl-1, whereas the three major proteasomal enzymatic activities were upregulated by palytoxin. Palytoxin-induced dephosphorylation of Bcl-2 further exacerbated the proapoptotic effect of Mcl-1 and Bcl-xL degradation in a range of leukemia cell lines. As okadaic acid rescued cell death triggered by palytoxin, protein phosphatase (PP)2A was involved in Bcl-2 dephosphorylation and induction of apoptosis by palytoxin. At a translational level, palytoxin abrogated the colony formation capacity of leukemia cell types. Moreover, palytoxin abrogated tumor formation in a zebrafish xenograft assay at concentrations between 10 and 30 pM. Altogether, we provide evidence of the role of palytoxin as a very potent and promising anti-leukemic agent, acting at low picomolar concentrations in cellulo and in vivo.
Collapse
Affiliation(s)
- Jaemyun Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
| | - Seungwon Ji
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
| | - Jin-Young Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
| | - Jean Lorquin
- Institut Méditerranéen d'Océanologie, 163 Avenue de Luminy, CEDEX 09, 13288 Marseille, France
| | - Barbora Orlikova-Boyer
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210 Luxembourg, Luxembourg
| | - Claudia Cerella
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210 Luxembourg, Luxembourg
| | - Aloran Mazumder
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
| | - Florian Muller
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210 Luxembourg, Luxembourg
| | - Olivier Detournay
- Planktovie SAS, 45 Rue Frédéric Joliot Curie, CEDEX 13, 13013 Marseille, France
| | - Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
| |
Collapse
|
47
|
Combination Therapies Targeting Apoptosis in Paediatric AML: Understanding the Molecular Mechanisms of AML Treatments Using Phosphoproteomics. Int J Mol Sci 2023; 24:ijms24065717. [PMID: 36982791 PMCID: PMC10058112 DOI: 10.3390/ijms24065717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
Paediatric acute myeloid leukaemia (AML) continues to present treatment challenges, as no “standard approach” exists to treat those young patients reliably and safely. Combination therapies could become a viable treatment option for treating young patients with AML, allowing multiple pathways to be targeted. Our in silico analysis of AML patients highlighted “cell death and survival” as an aberrant, potentially targetable pathway in paediatric AML patients. Therefore, we aimed to identify novel combination therapies to target apoptosis. Our apoptotic drug screening resulted in the identification of one potential “novel” drug pairing, comprising the Bcl-2 inhibitor ABT-737 combined with the CDK inhibitor Purvalanol-A, as well as one triple combination of ABT-737 + AKT inhibitor + SU9516, which showed significant synergism in a series of paediatric AML cell lines. Using a phosphoproteomic approach to understand the apoptotic mechanism involved, proteins related to apoptotic cell death and cell survival were represented, in agreement with further results showing differentially expressed apoptotic proteins and their phosphorylated forms among combination treatments compared to single-agent treated cells such upregulation of BAX and its phosphorylated form (Thr167), dephosphorylation of BAD (Ser 112), and downregulation of MCL-1 and its phosphorylated form (Ser159/Thr 163). Total levels of Bcl-2 were decreased but correlated with increased levels of phosphorylated Bcl-2, which was consistent with our phosphoproteomic analysis predictions. Bcl-2 phosphorylation was regulated by extracellular-signal-regulated kinase (ERK) but not PP2A phosphatase. Although the mechanism linking to Bcl-2 phosphorylation remains to be determined, our findings provide first-hand insights on potential novel combination treatments for AML.
Collapse
|
48
|
Kamolphiwong R, Kanokwiroon K, Wongrin W, Chaiyawat P, Klangjorhor J, Settakorn J, Teeyakasem P, Sangphukieo A, Pruksakorn D. Potential target identification for osteosarcoma treatment: Gene expression re-analysis and drug repurposing. Gene X 2023; 856:147106. [PMID: 36513192 DOI: 10.1016/j.gene.2022.147106] [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: 02/15/2022] [Revised: 11/18/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Survival rate of osteosarcoma has remained plateaued for the past three decades. New treatment is needed to improve survival rate. Drug repurposing, a method to identify new indications of previous drugs, which saves time and cost compared to the de novo drug discovery. Data mining from gene expression profile was carried out and new potential targets were identified by using drug repurposing strategy. Selected data were newly categorized as pathophysiology and metastasis groups. Data were normalized and calculated the differential gene expression. Genes with log fold change ≥ 2 and adjusted p-value ≤ 0.05 were selected as primary candidate genes (PCGs). PCGs were further enriched to determine the secondary candidate genes (SCGs) by protein interaction analysis, upstream transcription factor and related-protein kinase identification. PCGs and SCGs were further matched with gene targeted of corresponding drugs from the Drug Repurposing Hub. A total of 778 targets were identified (360 from PCGs, and 418 from SCGs). This newly identified KLHL13 is a new candidate target based on its molecular function. KLHL13 was upregulated in clinical samples. We found 256 drugs from matching processes (50anti-cancerand206non-anticancerdrugs). Clinical trials of anti-cancer drugs from 5 targets (CDK4, BCL-2, JUN, SRC, PIK3CA) are being performed for osteosarcoma treatment. Niclosamide and synthetic PPARɣ ligands are candidates for repurposing due to the possibility based on their mechanism and pharmacology properties. Re-analysis of gene expression profile could identify new potential targets, confirm a current implication, and expand the chance of repurposing drugs for osteosarcoma treatment.
Collapse
Affiliation(s)
- Rawikant Kamolphiwong
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Kanyanatt Kanokwiroon
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand.
| | - Weerinrada Wongrin
- Department of Statistics, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
| | - Parunya Chaiyawat
- Musculoskeletal Science and Translational Research Center, Department of Orthopaedics, Chiang Mai University, Chiang Mai, Thailand; Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Thailand
| | - Jeerawan Klangjorhor
- Musculoskeletal Science and Translational Research Center, Department of Orthopaedics, Chiang Mai University, Chiang Mai, Thailand; Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Thailand
| | - Jongkolnee Settakorn
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pimpisa Teeyakasem
- Musculoskeletal Science and Translational Research Center, Department of Orthopaedics, Chiang Mai University, Chiang Mai, Thailand
| | - Apiwat Sangphukieo
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Thailand
| | - Dumnoensun Pruksakorn
- Musculoskeletal Science and Translational Research Center, Department of Orthopaedics, Chiang Mai University, Chiang Mai, Thailand; Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Thailand.
| |
Collapse
|
49
|
Manai F, Zanoletti L, Morra G, Mansoor S, Carriero F, Bozzola E, Muscianisi S, Comincini S. Gluten Exorphins Promote Cell Proliferation through the Activation of Mitogenic and Pro-Survival Pathways. Int J Mol Sci 2023; 24:3912. [PMID: 36835317 PMCID: PMC9966116 DOI: 10.3390/ijms24043912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Celiac disease (CD) is a chronic and systemic autoimmune disorder that affects preferentially the small intestine of individuals with a genetic predisposition. CD is promoted by the ingestion of gluten, a storage protein contained in the endosperm of the seeds of wheat, barley, rye, and related cereals. Once in the gastrointestinal (GI) tract, gluten is enzymatically digested with the consequent release of immunomodulatory and cytotoxic peptides, i.e., 33mer and p31-43. In the late 1970s a new group of biologically active peptides, called gluten exorphins (GEs), was discovered and characterized. In particular, these short peptides showed a morphine-like activity and high affinity for the δ-opioid receptor (DOR). The relevance of GEs in the pathogenesis of CD is still unknown. Recently, it has been proposed that GEs could contribute to asymptomatic CD, which is characterized by the absence of symptoms that are typical of this disorder. In the present work, GEs cellular and molecular effects were in vitro investigated in SUP-T1 and Caco-2 cells, also comparing viability effects with human normal primary lymphocytes. As a result, GEs treatments increased tumor cell proliferation by cell cycle and Cyclins activation as well as by induction of mitogenic and pro-survival pathways. Finally, a computational model of GEs interaction with DOR is provided. Altogether, the results might suggest a possible role of GEs in CD pathogenesis and on its associated cancer comorbidities.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Lisa Zanoletti
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
- Laboratory for Mucosal Immunology, TARGID, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Giulia Morra
- SCITEC, Consiglio Nazionale delle Ricerche, 20131 Milano, Italy
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Samman Mansoor
- SCITEC, Consiglio Nazionale delle Ricerche, 20131 Milano, Italy
| | - Francesca Carriero
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Elena Bozzola
- Pediatric Unit, I.R.C.C.S. Bambino Gesù Children Hospital, 00165 Roma, Italy
| | - Stella Muscianisi
- Cell Factory and Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Sergio Comincini
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
50
|
Mafi A, Rezaee M, Hedayati N, Hogan SD, Reiter RJ, Aarabi MH, Asemi Z. Melatonin and 5-fluorouracil combination chemotherapy: opportunities and efficacy in cancer therapy. Cell Commun Signal 2023; 21:33. [PMID: 36759799 PMCID: PMC9912526 DOI: 10.1186/s12964-023-01047-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/14/2023] [Indexed: 02/11/2023] Open
Abstract
Combined chemotherapy is a treatment method based on the simultaneous use of two or more therapeutic agents; it is frequently necessary to produce a more effective treatment for cancer patients. Such combined treatments often improve the outcomes over that of the monotherapy approach, as the drugs synergistically target critical cell signaling pathways or work independently at different oncostatic sites. A better prognosis has been reported in patients treated with combination therapy than in patients treated with single drug chemotherapy. In recent decades, 5-fluorouracil (5-FU) has become one of the most widely used chemotherapy agents in cancer treatment. This medication, which is soluble in water, is used as the first line of anti-neoplastic agent in the treatment of several cancer types including breast, head and neck, stomach and colon cancer. Within the last three decades, many studies have investigated melatonin as an anti-cancer agent; this molecule exhibits various functions in controlling the behavior of cancer cells, such as inhibiting cell growth, inducing apoptosis, and inhibiting invasion. The aim of this review is to comprehensively evaluate the role of melatonin as a complementary agent with 5-FU-based chemotherapy for cancers. Additionally, we identify the potential common signaling pathways by which melatonin and 5-FU interact to enhance the efficacy of the combined therapy. Video abstract.
Collapse
Affiliation(s)
- Alireza Mafi
- grid.411036.10000 0001 1498 685XDepartment of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran
| | - Malihe Rezaee
- grid.411600.2School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran ,grid.411705.60000 0001 0166 0922Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Neda Hedayati
- grid.411746.10000 0004 4911 7066School of Medicine, Iran University of Medical Science, Tehran, Islamic Republic of Iran
| | - Sara Diana Hogan
- grid.8993.b0000 0004 1936 9457Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Russel J. Reiter
- grid.43582.380000 0000 9852 649XDepartment of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX USA
| | - Mohammad-Hossein Aarabi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|