1
|
Lin X, Deng S, Fu T, Lei Y, Wang Y, Yao J, Lu Y, Huang Y, Shang J, Chen J, Zhou X. Hyaluronic acid-based hydrogel microspheres with multi-responsive properties for antibacterial therapy and bone regeneration in Staphylococcus aureus-infected skull defects. Mater Today Bio 2025; 32:101676. [PMID: 40236808 PMCID: PMC11997343 DOI: 10.1016/j.mtbio.2025.101676] [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: 12/30/2024] [Revised: 02/20/2025] [Accepted: 03/16/2025] [Indexed: 04/17/2025] Open
Abstract
This study introduces hyaluronic acid-based (HA) hydrogel microspheres loaded with zinc oxide nanoparticles (ZnO-NPs) for the treatment of infectious bone defects. The microspheres were fabricated using a 3D-printing process, with a formulation consisting of 6 wt% HAD (methacrylated HA), 3 wt% AOHA (AMP-conjugated oxidized HA), 1 % BOHA (phenylboric acid-conjugated HA), 0.5 % photoinitiator, and 0.05 % ZnO-NPs. In vitro, the hydrogel microspheres demonstrated significant antibacterial activity against Staphylococcus aureus, with colony counts and biofilm inhibition assays showing a marked reduction in bacterial growth after 12 and 24 h. The release of antimicrobial peptides (AMPs) was enhanced in acidic conditions and in the presence of hyaluronidase. The microspheres also promoted osteogenic differentiation of bone marrow stromal cells (BMSCs), as evidenced by increased expression of osteogenic markers (ALP, OCN, OPN, and COL-1). In vivo, the hydrogel microspheres were tested in a rat skull defect model, showing significant bone regeneration, improved angiogenesis, and an anti-inflammatory response. These results indicate that ABOHA@ZnO hydrogel microspheres provide a promising strategy for treating infectious bone defects by combining antimicrobial, osteogenic.
Collapse
Affiliation(s)
- Xiaolong Lin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Shuli Deng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Tao Fu
- Department of Oral and Maxillofacial Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, School of Stomatology and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310000, China
| | - Yuqing Lei
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital, School of Biomedical Engineering Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Jiapei Yao
- Department of Orthopedics, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, 213000, China
| | - Yaojun Lu
- Department of Orthopedics, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, 213000, China
| | - Yong Huang
- Department of Orthopedics, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, 213000, China
| | - Jingjing Shang
- Department of Pharmacy, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, Jiangsu, 213000, China
| | - Jingjing Chen
- Department of Orthopedics, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, 213000, China
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, NHC Key Laboratory of Antibody Technique, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Xindie Zhou
- Department of Orthopedics, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, 213000, China
- Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture, Qinghai Province, 811800, China
| |
Collapse
|
2
|
Wu X, Yang Z, Zou J, Gao H, Shao Z, Li C, Lei P. Protein kinases in neurodegenerative diseases: current understandings and implications for drug discovery. Signal Transduct Target Ther 2025; 10:146. [PMID: 40328798 PMCID: PMC12056177 DOI: 10.1038/s41392-025-02179-x] [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: 11/01/2024] [Revised: 01/03/2025] [Accepted: 02/12/2025] [Indexed: 05/08/2025] Open
Abstract
Neurodegenerative diseases (e.g., Alzheimer's, Parkinson's, Huntington's disease, and Amyotrophic Lateral Sclerosis) are major health threats for the aging population and their prevalences continue to rise with the increasing of life expectancy. Although progress has been made, there is still a lack of effective cures to date, and an in-depth understanding of the molecular and cellular mechanisms of these neurodegenerative diseases is imperative for drug development. Protein phosphorylation, regulated by protein kinases and protein phosphatases, participates in most cellular events, whereas aberrant phosphorylation manifests as a main cause of diseases. As evidenced by pharmacological and pathological studies, protein kinases are proven to be promising therapeutic targets for various diseases, such as cancers, central nervous system disorders, and cardiovascular diseases. The mechanisms of protein phosphatases in pathophysiology have been extensively reviewed, but a systematic summary of the role of protein kinases in the nervous system is lacking. Here, we focus on the involvement of protein kinases in neurodegenerative diseases, by summarizing the current knowledge on the major kinases and related regulatory signal transduction pathways implicated in diseases. We further discuss the role and complexity of kinase-kinase networks in the pathogenesis of neurodegenerative diseases, illustrate the advances of clinical applications of protein kinase inhibitors or novel kinase-targeted therapeutic strategies (such as antisense oligonucleotides and gene therapy) for effective prevention and early intervention.
Collapse
Affiliation(s)
- Xiaolei Wu
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhangzhong Yang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinjun Zou
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Kiang JG, Cannon G, Zhai M, Olson MG, Woods AK, Cleveland KS, Ellery H, Xu F, Xiao M. A Combined Therapy of Pegylated G-CSF with Ciprofloxacin Mitigates Damage Induced by Lethal Ionizing Radiation to the Bone Marrow, Spleen, and Ileum by Increasing AKT Activation but Decreasing IL-18, C3, and miR-34a. Radiat Res 2025; 203:341-356. [PMID: 40181563 DOI: 10.1667/rade-24-00266.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Ciprofloxacin (CIP) was found to enhance pegylated G-CSF therapy (PEG, Neulasta®)-induced survival from 30% to 85% after ionizing radiation exposure. This combined therapy significantly mitigated radiation-induced brain hemorrhage through its capability to improve platelet recovery. This study tested whether this combined treatment also mitigated gastrointestinal damage from radiation. B6D2F1 female mice were exposed to 60Co γ radiation. CIP was fed daily to mice for up to 14 days. PEG was injected on day 1, and then weekly up to day 14. For the early time point study, blood, femurs, spleen, and ileum were collected on days 2, 4, 9, and 15 postirradiation. Bone marrow cells were counted; spleen weights and splenocyte counts were measured; and ileum histopathology was examined and analyzed. AKT, ERK, JNK, p38, claudin 2, NF-kB, Bax, Bcl-2, and gasdermin D were measured in ileum lysates using Western blotting while miR-34a was measured by reverse transcription followed by real-time-PCR, and citrulline was measured by colorimetric assay. In serum, interleukin-18 (IL-18) was measured by Luminex assay and complement protein 3 (C3) was detected by ELISA. The bacterial DNA load in livers was measured by real-time PCR. Radiation depleted bone marrow cells in femurs beginning day 2 through day 15 postirradiation, which was mitigated by PEG or CIP+PEG on day 9 through day 15 and by CIP on day 15, respectively. Radiation exposure led to decreased spleen weight on day 2 through day 15, while PEG or CIP+PEG significantly mitigated the reduction on day 9 through day 15. Radiation exposure reduced splenocyte counts on day 2 through day 15 postirradiation, but that was mitigated by PEG or CIP+PEG on day 15. Ileum histology showed that radiation decreased villus height on day 2 through day 15; CIP mitigated the reduction on day 15, whereas PEG+CIP mitigated it on day 2 through 15. Villus widths were increased on day 2 through day 15, while PEG+CIP effectively decreased them on day 4 through day 15. Crypt depth was reduced by radiation on day 2, but returned to the baseline on day 4 through 15. CIP or CIP+PEG transiently increased the depth only on day 4. Crypt counts were reduced by radiation on days 2 and 4, but returned to the baseline on days 9 and 15, regardless of individual drugs or combinations. Citrulline data confirmed the villus height recovery. Radiation significantly increased pro-inflammatory cytokine IL-18 on days 4 and 9, which was mitigated by PEG alone or PEG+CIP, but not by CIP alone. Radiation increased C3 on day 9 in ileum and serum. The serum C3 was positively associated with the serum IL-18 levels and negatively correlated with the crypt depth. Radiation-induced decreases in claudin 2 (a tight junction marker) in ileum and increases in bacterial DNA in livers were mitigated by PEG+CIP. Radiation did not reduce NF-kB and its activation but reduced Bcl-2 expression, which was not significantly recovered by any individual drug or combination. However, the PEG and CIP combination significantly decreased NF- kB and BAX. In contrast, radiation increased miR-34a and cleaved gasdermin D, which CIP+PEG effectively mitigated. This was confirmed by immunohistochemistry. The results taken together suggest that PEG+CIP combined treatment was effective in mitigating the radiation-induced bone marrow, spleen, and ileum injury. The mitigative effect of this combined treatment was mediated by increases in G-CSF levels that suppress miR-34a, thereby probably leading to decreased gasdermin D-mediated pyroptosis.
Collapse
Affiliation(s)
- Juliann G Kiang
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | - Georgetta Cannon
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Min Zhai
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Matthew G Olson
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Akeylah K Woods
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Katherine S Cleveland
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Hengying Ellery
- Department of Large Animal Research, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | - Feng Xu
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| | - Mang Xiao
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889
| |
Collapse
|
4
|
Hodges A, Dubuque R, Chen SH, Pan PY. The LILRB family in hematologic malignancies: prognostic associations, mechanistic considerations, and therapeutic implications. Biomark Res 2024; 12:159. [PMID: 39696628 DOI: 10.1186/s40364-024-00705-7] [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: 08/19/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
The leukocyte immunoglobulin-like receptor B (LILRB) proteins, characterized by their transmembrane nature and canonical immunoreceptor tyrosine-based inhibitory motifs (ITIM) signaling, play a pivotal role in maintaining immune homeostasis and are implicated in the pathogenesis of various disease states. This comprehensive review will focus on the intricate involvement of the LILRB family in hematologic malignancies. These receptors have emerged as valuable diagnostic and prognostic biomarkers in leukemia, lymphoma, and myeloma. Beyond their prognostic implications, LILRBs actively shape the immune microenvironment and directly influence the disease pathogenesis of hematologic malignancies. Furthermore, their identification as potential therapeutic targets offer a promising avenue for precision medicine strategies in the treatment of these disorders. Currently, multiple LILRB directed therapies are in the preclinical and clinical trial pipelines. This review underscores the multifaceted role of the LILRB family in hematologic malignancies, highlighting their significance from diagnostic and prognostic perspectives to their broader impact on disease pathophysiology and as valuable therapeutic targets.
Collapse
Affiliation(s)
- Alan Hodges
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Texas A&M University College of Medicine, Bryan, TX, 77807, USA
| | - Rachel Dubuque
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medical Science and Graduate School of Medical Sciences, New York City, NY, 10065, USA
| | - Shu-Hsia Chen
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Texas A&M University College of Medicine, Bryan, TX, 77807, USA.
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medical Science and Graduate School of Medical Sciences, New York City, NY, 10065, USA.
| | - Ping-Ying Pan
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Texas A&M University College of Medicine, Bryan, TX, 77807, USA.
| |
Collapse
|
5
|
Yang C, da Silva MCM, Howell JA, Larochelle J, Liu L, Gunraj RE, de Oliveira ACP, Candelario-Jalil E. RIPK2 Is Crucial for the Microglial Inflammatory Response to Bacterial Muramyl Dipeptide but Not to Lipopolysaccharide. Int J Mol Sci 2024; 25:11754. [PMID: 39519307 PMCID: PMC11546996 DOI: 10.3390/ijms252111754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Receptor-interacting serine/threonine protein kinase 2 (RIPK2) is a kinase that is essential in modulating innate and adaptive immune responses. As a downstream signaling molecule for nucleotide-binding oligomerization domain 1 (NOD1), NOD2, and Toll-like receptors (TLRs), it is implicated in the signaling triggered by recognition of microbe-associated molecular patterns by NOD1/2 and TLRs. Upon activation of these innate immune receptors, RIPK2 mediates the release of pro-inflammatory factors by activating mitogen-activated protein kinases (MAPKs) and nuclear factor-kappa B (NF-κB). However, whether RIPK2 is essential for downstream inflammatory signaling following the activation of NOD1/2, TLRs, or both remains controversial. In this study, we examined the role of RIPK2 in NOD2- and TLR4-dependent signaling cascades following stimulation of microglial cells with bacterial muramyl dipeptide (MDP), a NOD2 agonist, or lipopolysaccharide (LPS), a TLR4 agonist. We utilized a highly specific proteolysis targeting chimera (PROTAC) molecule, GSK3728857A, and found dramatic degradation of RIPK2 in a concentration- and time-dependent manner. Importantly, the PROTAC completely abolished MDP-induced increases in iNOS and COX-2 protein levels and pro-inflammatory gene transcription of Nos2, Ptgs2, Il-1β, Tnfα, Il6, Ccl2, and Mmp9. However, increases in iNOS and COX-2 proteins and pro-inflammatory gene transcription induced by the TLR4 agonist, LPS, were only slightly attenuated with the GSK3728857A pretreatment. Further findings revealed that the RIPK2 PROTAC completely blocked the phosphorylation and activation of p65 NF-κB and p38 MAPK induced by MDP, but it had no effects on the phosphorylation of these two mediators triggered by LPS. Collectively, our findings strongly suggest that RIPK2 plays an essential role in the inflammatory responses of microglia to bacterial MDP but not to LPS.
Collapse
Affiliation(s)
- Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL 32610, USA; (C.Y.); (M.C.M.d.S.); (J.A.H.); (J.L.); (L.L.); (R.E.G.); (A.C.P.d.O.)
| | - Maria Carolina Machado da Silva
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL 32610, USA; (C.Y.); (M.C.M.d.S.); (J.A.H.); (J.L.); (L.L.); (R.E.G.); (A.C.P.d.O.)
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - John Aaron Howell
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL 32610, USA; (C.Y.); (M.C.M.d.S.); (J.A.H.); (J.L.); (L.L.); (R.E.G.); (A.C.P.d.O.)
| | - Jonathan Larochelle
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL 32610, USA; (C.Y.); (M.C.M.d.S.); (J.A.H.); (J.L.); (L.L.); (R.E.G.); (A.C.P.d.O.)
| | - Lei Liu
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL 32610, USA; (C.Y.); (M.C.M.d.S.); (J.A.H.); (J.L.); (L.L.); (R.E.G.); (A.C.P.d.O.)
| | - Rachel E. Gunraj
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL 32610, USA; (C.Y.); (M.C.M.d.S.); (J.A.H.); (J.L.); (L.L.); (R.E.G.); (A.C.P.d.O.)
| | - Antônio Carlos Pinheiro de Oliveira
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL 32610, USA; (C.Y.); (M.C.M.d.S.); (J.A.H.); (J.L.); (L.L.); (R.E.G.); (A.C.P.d.O.)
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL 32610, USA; (C.Y.); (M.C.M.d.S.); (J.A.H.); (J.L.); (L.L.); (R.E.G.); (A.C.P.d.O.)
| |
Collapse
|
6
|
Li J, Chen K, Li X, Zhang X, Zhang L, Yang Q, Xia Y, Xie C, Wang X, Tong J, Shen Y. Mechanistic insights into the alterations and regulation of the AKT signaling pathway in diabetic retinopathy. Cell Death Discov 2023; 9:418. [PMID: 37978169 PMCID: PMC10656479 DOI: 10.1038/s41420-023-01717-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
In the early stages of diabetic retinopathy (DR), diabetes-related hyperglycemia directly inhibits the AKT signaling pathway by increasing oxidative stress or inhibiting growth factor expression, which leads to retinal cell apoptosis, nerve proliferation and fundus microvascular disease. However, due to compensatory vascular hyperplasia in the late stage of DR, the vascular endothelial growth factor (VEGF)/phosphatidylinositol 3 kinase (PI3K)/AKT cascade is activated, resulting in opposite levels of AKT regulation compared with the early stage. Studies have shown that many factors, including insulin, insulin-like growth factor-1 (IGF-1), VEGF and others, can regulate the AKT pathway. Disruption of the insulin pathway decreases AKT activation. IGF-1 downregulation decreases the activation of AKT in DR, which abrogates the neuroprotective effect, upregulates VEGF expression and thus induces neovascularization. Although inhibiting VEGF is the main treatment for neovascularization in DR, excessive inhibition may lead to apoptosis in inner retinal neurons. AKT pathway substrates, including mammalian target of rapamycin (mTOR), forkhead box O (FOXO), glycogen synthase kinase-3 (GSK-3)/nuclear factor erythroid 2-related factor 2 (Nrf2), and nuclear factor kappa-B (NF-κB), are a research focus. mTOR inhibitors can delay or prevent retinal microangiopathy, whereas low mTOR activity can decrease retinal protein synthesis. Inactivated AKT fails to inhibit FOXO and thus causes apoptosis. The GSK-3/Nrf2 cascade regulates oxidation and inflammation in DR. NF-κB is activated in diabetic retinas and is involved in inflammation and apoptosis. Many pathways or vital activities, such as the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) and mitogen-activated protein kinase (MAPK) signaling pathways, interact with the AKT pathway to influence DR development. Numerous regulatory methods can simultaneously impact the AKT pathway and other pathways, and it is essential to consider both the connections and interactions between these pathways. In this review, we summarize changes in the AKT signaling pathway in DR and targeted drugs based on these potential sites.
Collapse
Affiliation(s)
- Jiayuan Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kuangqi Chen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiang Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xuhong Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Liyue Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Qianjie Yang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yutong Xia
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Chen Xie
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiawei Wang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianping Tong
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China.
| | - Ye Shen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Meng F, Zhou Y, Wagner A, Bülow JM, Köhler K, Neunaber C, Bundkirchen K, Relja B. Impact of age on liver damage, inflammation, and molecular signaling pathways in response to femoral fracture and hemorrhage. Front Immunol 2023; 14:1239145. [PMID: 37691959 PMCID: PMC10484338 DOI: 10.3389/fimmu.2023.1239145] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Background Trauma causes disability and mortality globally, leading to fractures and hemorrhagic shock. This can trigger an irregular inflammatory response that damages remote organs, including liver. Aging increases the susceptibility to dysregulated immune responses following trauma, raising the risk of organ damage, infections, and higher morbidity and mortality in elderly patients. This study investigates how aging affects liver inflammation and damage post-trauma. Methods 24 male C57BL/6J mice were randomly divided into four groups. Twelve young (17-26 weeks) and 12 aged (64-72 weeks) mice were included. Mice further underwent either hemorrhagic shock (trauma/hemorrhage, TH), and femoral fracture (osteotomy) with external fixation (Fx) (THFx, n=6) or sham procedures (n=6). After 24 hours, mice were sacrificed. Liver injury and apoptosis were evaluated using hematoxylin-eosin staining and activated caspase-3 immunostaining. CXCL1 and infiltrating polymorphonuclear leukocytes (PMNL) in the liver were assessed by immunostaining, and concentrations of CXCL1, TNF, IL-1β, and IL-10 in the liver tissue were determined by ELISA. Gene expression of Tnf, Cxcl1, Il-1β, and Cxcl2 in the liver tissue was determined by qRT-PCR. Finally, western blot was used to determine protein expression levels of IκBα, Akt, and their phosphorylated forms. Results THFx caused liver damage and increased presence of active caspase-3-positive cells compared to the corresponding sham group. THFx aged group had more severe liver injury than the young group. CXCL1 and PMNL levels were significantly higher in both aged groups, and THFx caused a greater increase in CXCL and PMNL levels in aged compared to the young group. Pro-inflammatory TNF and IL-1β levels were elevated in aged groups, further intensified by THFx. Anti-inflammatory IL-10 levels were lower in aged groups. Tnf and Cxcl1 gene expression was enhanced in the aged sham group. Phosphorylation ratio of IκBα was significantly increased in the aged sham group versus young sham group. THFx-induced IκBα phosphorylation in the young group was significantly reduced in the aged THFx group. Akt phosphorylation was significantly reduced in the THFx aged group compared to the THFx young group. Conclusion The findings indicate that aging may lead to increased vulnerability to liver injury and inflammation following trauma due to dysregulated immune responses.
Collapse
Affiliation(s)
- Fanshuai Meng
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
- Department of Trauma and Reconstructive Surgery, Uniklinik RWTH Aachen, Aachen, Germany
| | - Yuzhuo Zhou
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Alessa Wagner
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
| | - Jasmin Maria Bülow
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
| | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Claudia Neunaber
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Katrin Bundkirchen
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Borna Relja
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
8
|
Kuczyńska M, Gabig-Cimińska M, Moskot M. Molecular treatment trajectories within psoriatic T lymphocytes: a mini review. Front Immunol 2023; 14:1170273. [PMID: 37251381 PMCID: PMC10213638 DOI: 10.3389/fimmu.2023.1170273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
Multiple biological processes in mammalian cells are implicated in psoriasis (Ps) development and progression, as well as in the pathogenic mechanisms associated with this chronic immune-mediated inflammatory disease (IMID). These refer to molecular cascades contributing to the pathological topical and systemic reactions in Ps, where local skin-resident cells derived from peripheral blood and skin-infiltrating cells originating from the circulatory system, in particular T lymphocytes (T cells), are key actors. The interplay between molecular components of T cell signalling transduction and their involvement in cellular cascades (i.e. throughout Ca2+/CaN/NFAT, MAPK/JNK, PI3K/Akt/mTOR, JAK/STAT pathways) has been of concern in the last few years; this is still less characterised than expected, even though some evidence has accumulated to date identifying them as potential objects in the management of Ps. Innovative therapeutic strategies for the use of compounds such as synthetic Small Molecule Drugs (SMDs) and their various combinations proved to be promising tools for the treatment of Ps via incomplete blocking, also known as modulation of disease-associated molecular tracks. Despite recent drug development having mainly centred on biological therapies for Ps, yet displaying serious limitations, SMDs acting on specific pathway factor isoforms or single effectors within T cell, could represent a valid innovation in real-world treatment patterns in patients with Ps. Of note, due to the intricate crosstalk between intracellular pathways, the use of selective agents targeting proper tracks is, in our opinion, a challenge for modern science regarding the prevention of disease at its onset and also in the prediction of patient response to Ps treatment.
Collapse
Affiliation(s)
| | | | - Marta Moskot
- Department of Medical Biology and Genetics, University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
9
|
Li C, Wang S, Ma X, Wang T, Lu R, Jia X, Leng Z, Kong X, Zhang J, Li L. Ranitidine as an adjuvant regulates macrophage polarization and activates CTLs through the PI3K-Akt2 signaling pathway. Int Immunopharmacol 2023; 116:109729. [PMID: 37800555 DOI: 10.1016/j.intimp.2023.109729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 02/19/2023]
Abstract
Adjuvants are an indispensable component of vaccines, but there are few adjuvants for human vaccines. H2 receptor blockers, inhibiting gastric acid secretion, have immune enhancement effects. Ranitidine (RAN) is a water-soluble H2 receptor blocker, and whether it has an immune-enhancing effect is still unknown. In this study, flow cytometry, western blotting, and immunofluorescence methods were used to analyze whether RAN could activate macrophage polarization to the M1 phenotype in vivo and in vitro. Here, we found that the M1 inflammatory cytokine levels and surface markers in RAW264.7 cells were upregulated by NF-κB activation, possibly through the PI3K-Akt2 signaling pathway, after RAN treatment. Endocytic function was also enhanced by feedback regulation of Akt2/GSK3β/Dynmin1 signaling. Furthermore, to evaluate the adjuvant function of RAN, we used OVA plus RAN as a vaccine to inhibit the growth of B16-OVA tumors in mice. We also found that in the RAN adjuvant group, macrophage polarization to M1, Th1 cell differentiation, and cytotoxic T lymphocyte (CTL) activation were significantly upregulated. The tumor growth of mice was inhibited, and the survival rate of mice was significantly improved. This study provides new evidence for the mechanism by which RAN activates the immune response and is expected to provide a new strategy for the research and development of tumor vaccine adjuvants.
Collapse
Affiliation(s)
- Chenglin Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Shuang Wang
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China.
| | - Xiaoran Ma
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Tiantian Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Ran Lu
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Xihui Jia
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Zhe Leng
- Department of Gynecology, Qingdao Women and Children's Hospital, Qingdao 266000, China
| | - Xiaowen Kong
- School of Stomatology, Qingdao University, Qingdao 266071, China
| | - Jinyu Zhang
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Ling Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China.
| |
Collapse
|
10
|
Osteopontin Splicing Isoforms Contribute to Endometriotic Proliferation, Migration, and Epithelial-Mesenchymal Transition in Endometrial Epithelial Cells. Int J Mol Sci 2022; 23:ijms232315328. [PMID: 36499654 PMCID: PMC9738877 DOI: 10.3390/ijms232315328] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Osteopontin (OPN) isoforms, including OPNb and OPNc, promote malignancy and may contribute to the pathogenesis of endometriosis, a benign disorder with multiple characteristics resembling malignant tumors. In our experiments, OPNb and OPNc were significantly overexpressed in both endometriosis and adenomyosis compared to the normal endometrium. Upregulation of CD44v and the epithelial-mesenchymal transition (EMT) process was also present in endometriotic lesions. Overexpression of OPNb and OPNc splicing variants in endometriotic cells evoked morphological changes, actin remodeling, cell proliferation, cell migration, and EMT through binding OPN ligand receptors CD44 and αvβ3, subsequently activating the PI3K and NF-ĸB pathways. We elucidated the causal role of OPN splice variants in regulating endometriotic cell growth, which may promote the development of OPN-targeted therapies for patients suffering from endometriotic disorders.
Collapse
|
11
|
Kırça M. Methylglyoxal enhances the proliferation of vascular smooth muscle cells via Akt phosphorylation. J Recept Signal Transduct Res 2022; 42:567-572. [PMID: 35818345 DOI: 10.1080/10799893.2022.2098328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Methylglyoxal (MGO) is predominantly produced as a by-product of the glycolysis pathway. The glyoxalase system effectively removes it in a healthy organism. However, this process is impaired, and MGO level is elevated in people with diabetes. MGO's effects on proliferation were mostly studied in cancer cells, and the data in other cell types are limited. This study inspected the proliferative capacity of MGO in vascular smooth muscle cells (VSMCs), which have a crucial role in atherosclerosis and restenosis. The roles of ERK1/2 MAPK and Akt phosphorylations in proliferation were determined. Telmisartan, irbesartan, and NF-κB inhibitor JSH-23's roles in protecting the cells from MGO-induced proliferation were also investigated. Primary VSMCs were isolated from the rat aorta. The proliferation was spectrophotometrically measured by using a tetrazolium salt (Wst-1). The cells were cultured in standard media (SM, glucose conc. 5.5 mM) or high glucose media (HGM, glucose conc. 25 mM; an in vitro model of hyperglycemia). ERK1/2 MAPK and Akt phosphorylations were determined by the western blot method. MGO triggered the proliferation at 24, 48, and 72 hrs in SM and 48 and 72 hrs in HGM. Low doses of MGO such as 1-10 µM can induce proliferation. The phosphorylated ERK1/2 MAPK and Akt participated in MGO-induced proliferation. Telmisartan, irbesartan, and JSH-23 effectively alleviated the proliferation and Akt phosphorylation. MGO could proliferate VSMCs even at low doses. Moreover, hypertensive diabetic patients might benefit from a sartan family drug to protect VSMCs from MGO-induced proliferation.
Collapse
Affiliation(s)
- Mustafa Kırça
- Department of Biochemistry, Faculty of Medicine, Kütahya Health Sciences University, Kütahya, Turkey
| |
Collapse
|
12
|
Walter LO, Maioral MF, Silva LO, Speer DB, Campbell SC, Gallimore W, Falkenberg MB, Santos-Silva MC. Involvement of the NF-κB and PI3K/Akt/mTOR pathways in cell death triggered by stypoldione, an o-quinone isolated from the brown algae Stypopodium zonale. ENVIRONMENTAL TOXICOLOGY 2022; 37:1297-1309. [PMID: 35128807 DOI: 10.1002/tox.23484] [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: 08/21/2021] [Revised: 01/07/2022] [Accepted: 01/22/2022] [Indexed: 06/14/2023]
Abstract
Multiple myeloma (MM) is a clonal plasma cell malignancy that remains incurable to date. Thus, the aims of this study were to evaluate the involvement of the NF-κB and PI3K/Akt/mTOR pathways in the cytotoxicity of stypoldione, an o-quinone isolated from the brown algae Stypopodium zonale, in MM cells (MM1.S). The cytotoxic effect was evaluated in MM1.S cells and peripheral blood mononuclear cells (PBMCs) by MTT assay. The stypoldione reduced the cell viability of MM1.S cells in a concentration and time-dependent manner (IC50 in MM.1S from 2.55 to 5.38 μM). However, it was also cytotoxic to PBMCs, but at a lower range. Additionally, no significant hemolysis was observed even at concentration up to 10 times the IC50 . Apoptotic cell death was confirmed by cell morphology and Annexin V-FITC assay. Stypoldione induced intrinsic and extrinsic apoptosis by increasing FasR expression and reactive oxygen species (ROS) production, inverting the Bax/Bcl-2 ratio, and inducing ΔΨm loss, which resulted in AIF release and caspase-3 activation. It also increased Ki-67 and survivin expression and inhibited the NF-κB and PI3K/Akt/mTOR pathways. These results suggest that stypoldione is a good candidate for the development of new drugs for MM treatment.
Collapse
Affiliation(s)
- Laura O Walter
- Experimental Oncology and Hemopathies Laboratory, Clinical Analysis Department, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
- Post-Graduation Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Mariana F Maioral
- Experimental Oncology and Hemopathies Laboratory, Clinical Analysis Department, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
- Post-Graduation Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Lisandra O Silva
- Experimental Oncology and Hemopathies Laboratory, Clinical Analysis Department, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
- Post-Graduation Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Douglas B Speer
- Experimental Oncology and Hemopathies Laboratory, Clinical Analysis Department, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Sanjay C Campbell
- Department of Chemistry, University of the West Indies, St. Andrew, Jamaica
| | - Winklet Gallimore
- Department of Chemistry, University of the West Indies, St. Andrew, Jamaica
| | - Miriam B Falkenberg
- Post-Graduation Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Maria Cláudia Santos-Silva
- Experimental Oncology and Hemopathies Laboratory, Clinical Analysis Department, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
- Post-Graduation Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
13
|
Vageli DP, Doukas SG, Doukas PG, Judson BL. Bile reflux and hypopharyngeal cancer (Review). Oncol Rep 2021; 46:244. [PMID: 34558652 PMCID: PMC8485019 DOI: 10.3892/or.2021.8195] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/24/2021] [Indexed: 12/26/2022] Open
Abstract
Laryngopharyngeal reflux, a variant of gastroesophageal reflux disease, has been considered a risk factor in the development of hypopharyngeal cancer. Bile acids are frequently present in the gastroesophageal refluxate and their effect has been associated with inflammatory and neoplastic changes in the upper aerodigestive tract. Recent in vitro and in vivo studies have provided direct evidence of the role of acidic bile refluxate in hypopharyngeal carcinogenesis and documented the crucial role of NF-κB as a key mediator of early oncogenic molecular events in this process and also suggested a contribution of STAT3. Acidic bile can cause premalignant changes and invasive squamous cell cancer in the affected hypopharynx accompanied by DNA damage, elevated p53 expression and oncogenic mRNA and microRNA alterations, previously linked to head and neck cancer. Weakly acidic bile can also increase the risk for hypopharyngeal carcinogenesis by inducing DNA damage, exerting anti-apoptotic effects and causing precancerous lesions. The most important findings that strongly support bile reflux as an independent risk factor for hypopharyngeal cancer are presented in the current review and the underlying mechanisms are provided.
Collapse
Affiliation(s)
- Dimitra P Vageli
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sotirios G Doukas
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Panagiotis G Doukas
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Benjamin L Judson
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
14
|
Li M, Li H, Yang S, Liao X, Zhao C, Wang F. MicroRNA-29b participates in the epithelial-mesenchymal transition of retinal pigment epithelial cells through p-p65. Exp Ther Med 2021; 22:868. [PMID: 34194546 DOI: 10.3892/etm.2021.10300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 05/10/2021] [Indexed: 11/05/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is considered to be the main mechanism of proliferative vitreoretinopathy (PVR). Our previous study demonstrated that microRNA-29b (miR-29b) and its target protein kinase B (Akt2) played vital roles in this process. miR-29b, a mesenchymal marker α-smooth muscle actin (α-SMA) and the epithelial marker E-cadherin were assessed in epiretinal membranes of patients with PVR. The potential mechanism of miR-29b and EMT was also investigated. The expression levels of miR-29b, E-cadherin, and α-SMA in PVR epiretinal membranes were measured using quantitative PCR. The expression levels of Akt2, phosphorylated (p)-Akt2, p65, p-p65, and Snail in ARPE-19 cells were assessed using western blotting. The expression levels of miR-29b were positively correlated with E-cadherin mRNA expression, while an inverse correlation was observed between miR-29b and α-SMA mRNA expression in epiretinal membranes of patients with PVR. When miR-29b was transfected into ARPE-19 cells, the expression levels of Akt2, p-Akt2, p-p65 and Snail were downregulated. shRNA-Akt2 inhibited p-p65 and Snail expression, while the NF-κB inhibitor BAY11-7082 reduced Snail expression. The Akt2/p-p65/Snail pathway may be the underlying mechanism of miR-29b in EMT of RPE cells. The results of the present study may provide a new strategy for prevention and therapy of PVR.
Collapse
Affiliation(s)
- Min Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Shanghai 200072, P.R. China
| | - Hui Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Shanghai 200072, P.R. China
| | - Shuai Yang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Shanghai 200072, P.R. China
| | - Xin Liao
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Shanghai 200072, P.R. China
| | - Chun Zhao
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Shanghai 200072, P.R. China
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Shanghai 200072, P.R. China
| |
Collapse
|
15
|
Vallée A, Vallée JN, Le Blanche A, Lecarpentier Y. PPARγ Agonists: Emergent Therapy in Endometriosis. Pharmaceuticals (Basel) 2021; 14:ph14060543. [PMID: 34204039 PMCID: PMC8229142 DOI: 10.3390/ph14060543] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/31/2021] [Accepted: 06/04/2021] [Indexed: 01/01/2023] Open
Abstract
Endometriosis is one of the major gynecological diseases of reproductive-age women. This disease is characterized by the presence of glands and stroma outside the uterine cavity. Several studies have shown the major role of inflammation, angiogenesis, adhesion and invasion, and apoptosis in endometriotic lesions. Nevertheless, the mechanisms underlying endometriotic mechanisms still remain unclear and therapies are not currently efficient. The introduction of new agents can be effective by improving the condition of patients. PPARγ ligands can directly modulate these pathways in endometriosis. However, data in humans remain low. Thus, the purpose of this review is to summarize the potential actions of PPARγ agonists in endometriosis by acting on inflammation, angiogenesis, invasion, adhesion, and apoptosis.
Collapse
Affiliation(s)
- Alexandre Vallée
- Department of Clinical Research and Innovation (DRCI), Foch Hospital, 92150 Suresnes, France
- Correspondence:
| | - Jean-Noël Vallée
- Centre Hospitalier Universitaire (CHU) Amiens Picardie, Université Picardie Jules Verne (UPJV), 80000 Amiens, France;
- DACTIM-Mis, Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, 86000 Poitiers, France
| | - Alain Le Blanche
- Laboratoire CeRSM (EA-2931), UPL, Université Paris Nanterre, F92000 Nanterre, France;
- Hôpital René-Dubos de Pontoise and Université de Versailles-Saint-Quentin, Simone Veil UFR des Sciences de la Santé, 78180 Montigny-le-Bretonneux, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien (GHEF), 77100 Meaux, France;
| |
Collapse
|
16
|
Chen R, Tsai J, Thompson PA, Chen Y, Xiong P, Liu C, Burrows F, Sivina M, Burger JA, Keating MJ, Wierda WG, Plunkett W. The multi-kinase inhibitor TG02 induces apoptosis and blocks B-cell receptor signaling in chronic lymphocytic leukemia through dual mechanisms of action. Blood Cancer J 2021; 11:57. [PMID: 33714981 PMCID: PMC7956145 DOI: 10.1038/s41408-021-00436-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/13/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
The constitutive activation of B-cell receptor (BCR) signaling, together with the overexpression of the Bcl-2 family anti-apoptotic proteins, represents two hallmarks of chronic lymphocytic leukemia (CLL) that drive leukemia cell proliferation and sustain their survival. TG02 is a small molecule multi-kinase inhibitor that simultaneously targets both of these facets of CLL pathogenesis. First, its inhibition of cyclin-dependent kinase 9 blocked the activation of RNA polymerase II and transcription. This led to the depletion of Mcl-1 and rapid induction of apoptosis in the primary CLL cells. This mechanism of apoptosis was independent of CLL prognostic factors or prior treatment history, but dependent on the expression of BAX and BAK. Second, TG02, which inhibits the members of the BCR signaling pathway such as Lck and Fyn, blocked BCR-crosslinking-induced activation of NF-κB and Akt, indicating abrogation of BCR signaling. Finally, the combination of TG02 and ibrutinib demonstrated moderate synergy, suggesting a future combination of TG02 with ibrutinib, or use in patients that are refractory to the BCR antagonists. Thus, the dual inhibitory activity on both the CLL survival pathway and BCR signaling identifies TG02 as a unique compound for clinical development in CLL and possibly other B cell malignancies.
Collapse
Affiliation(s)
- Rong Chen
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| | - Jennifer Tsai
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Department of Emergency Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Philip A Thompson
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Yuling Chen
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Ping Xiong
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Chaomei Liu
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Francis Burrows
- Tragara Pharmaceuticals, Carlsbad, CA, USA.,Kura Oncology, Inc., San Diego, CA, USA
| | - Mariela Sivina
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jan A Burger
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Michael J Keating
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - William G Wierda
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - William Plunkett
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
17
|
Lopez-Bergami P, Barbero G. The emerging role of Wnt5a in the promotion of a pro-inflammatory and immunosuppressive tumor microenvironment. Cancer Metastasis Rev 2021; 39:933-952. [PMID: 32435939 DOI: 10.1007/s10555-020-09878-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Wnt5a is the prototypical activator of the non-canonical Wnt pathways, and its overexpression has been implicated in the progression of several tumor types by promoting cell motility, invasion, EMT, and metastasis. Recent evidences have revealed a novel role of Wnt5a in the phosphorylation of the NF-κB subunit p65 and the activation of the NF-κB pathway in cancer cells. In this article, we review the molecular mechanisms and mediators defining a Wnt5a/NF-κB signaling pathway and propose that the aberrant expression of Wnt5a in some tumors drives a Wnt5a/NF-κB/IL-6/STAT3 positive feedback loop that amplifies the effects of Wnt5a. The evidences discussed here suggest that Wnt5a has a double effect on the tumor microenvironment. First, it activates an autocrine ROR1/Akt/p65 pathway that promotes inflammation and chemotaxis of immune cells. Then, Wnt5a activates a TLR/MyD88/p50 pathway exclusively in myelomonocytic cells promoting the synthesis of the anti-inflammatory cytokine IL-10 and a tolerogenic phenotype. As a result of these mechanisms, Wnt5a plays a negative role on immune cell function that contributes to an immunosuppressive tumor microenvironment and would contribute to resistance to immunotherapy. Finally, we summarized the development of different strategies targeting either Wnt5a or the Wnt5a receptor ROR1 that can be helpful for cancer therapy by contributing to generate a more immunostimulatory tumor microenvironment.
Collapse
Affiliation(s)
- Pablo Lopez-Bergami
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimonides, Hidalgo 775, Buenos Aires, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Gastón Barbero
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimonides, Hidalgo 775, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
18
|
Jiao Y, Zhao D, Gao F, Hu X, Hu X, Li M, Cui Y, Wei X, Xie C, Zhao Y, Gao Y. MicroRNA-520c-3p suppresses vascular endothelium dysfunction by targeting RELA and regulating the AKT and NF-κB signaling pathways. J Physiol Biochem 2021; 77:47-61. [PMID: 33411212 DOI: 10.1007/s13105-020-00779-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022]
Abstract
Endothelial injury, which can cause endothelial inflammation and dysfunction, is an important mechanism for the development of atherosclerotic plaque. This study aims to investigate the functional role of miR-520c-3p in vascular endothelium during inflammatory diseases such as atherosclerosis. Quantitative real-time PCR was used to detect miR-520c-3p expression in in human umbilical vein endothelial cells (HUVECs) after treatment with platelet-derived growth factor (PDGF). Furthermore, the effects of miR-520c-3p overexpression and silencing on cell proliferation, adhesion, and apoptosis were assessed. Bioinformatics analysis and Biotin-labeled miRNA pull-down assay were used to confirm the targets of miR-520-3p. Then, the effects of miR-520c-3p on AKT and NF-κB signaling pathways were detected by western blot. Herein, we observed that the expression level of miR-520c-3p was downregulated in HUVECs under PDGF stimulation. Overexpression of miR-520c-3p not only decreased cell adhesion but also promoted proliferation and inhibited apoptosis to protect the viability of endothelial cells. It was confirmed that RELA is the target of miR-520c-3p. MiR-520c-3p inhibited the protein phosphorylation of AKT and RELA, and si-RELA reversed the promotion of AKT and RELA protein phosphorylation by anti-miR-520c-3p. In summary, our study suggested that miRNA-520c-3p targeting RELA through AKT and NF-κB signaling pathways regulated the proliferation, apoptosis, and adhesion of vascular endothelial cells. We conclude that miR-520c-3p may play an important role in the suppression of endothelial injury, which could serve as a biomarker and therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Yan Jiao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Dandan Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Fuhua Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoyan Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xinxin Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Mei Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Cui
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoqing Wei
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ce Xie
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Zhao
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China.
- Molecular Medicine Laboratory, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| | - Ying Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China.
- Molecular Medicine Laboratory, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, China.
| |
Collapse
|
19
|
Song C, Jeong D, Hong YH, Li WY, Lee SW, Hossain MA, Taamalli A, Kim JH, Kim JH, Cho JY. Anti-Inflammatory and Photoaging-Protective Effects of Olea europaea through Inhibition of AP-1 and NF-
κ
B Pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:1895-1913. [PMID: 33308098 DOI: 10.1142/s0192415x20500950] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Olea europaea is a beneficial edible plant with a number of biological activities like anti-inflammatory, anti-oxidant, antithrombic, antihyperglycemic, and anti-ischemic activities. The mechanisms behind the antiphotoaging and anti-inflammatory effects of Olea europaea are not fully understood. To investigate how an ethanol extract of Olea europaea (Oe-EE) exerts these effects, we explored its activities in human keratinocytes and dermal fibroblasts. We assessed the anti-oxidant effects of Oe-EE via 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2′ -azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt (ABTS) assays and measured the expression levels of matrix metalloproteinases (MMPs), cyclooxygenase-2, interleukin (IL)-6, tumor necrosis factor (TNF)-α , and moisturizing factors. Antiphotoaging and anti-inflammatory mechanisms of Oe-EE were explored by assessing signaling molecule activation via immunoblotting. Oe-EE treatment decreased the mRNA expression level of MMPs, cyclooxygenase-2, IL-6, and TNF-α and restored type I collagen, filaggrin, and sirtuin 1 expression in UVB-irradiated cells. Furthermore, Oe-EE inhibited the activities of several activator protein 1 regulatory enzymes, including extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK), and inhibited nuclear factor (NF)-κ B pathway signaling proteins. Therefore, our results indicate that Oe-EE has photoaging-protective and anti-inflammatory effects.
Collapse
Affiliation(s)
- Chaoran Song
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Deok Jeong
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yo Han Hong
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Wan Yi Li
- Institute of Medicinal Plants, Yunnan Academy of Agricultural Sciences, Yunnan 650205, P. R. China
| | - Sang Woo Lee
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141 Republic of Korea
| | - Mohammad Amjad Hossain
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Amani Taamalli
- Laboratory of Olive Biotechnology, Center of Biotechnology-Technopole of Borj-Cedria, BP 901, Hammam-Lif 2050, Tunisia
- Department of Chemistry, University of Hafr Al Batin, Hafr Al Batin 31991, Kingdom of Saudi Arabia
| | - Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jong-Hoon Kim
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141 Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
20
|
Wang Y, Chen M, Xu J, Liu X, Duan Y, Zhou C, Xu Y. Core clock gene Bmal1 deprivation impairs steroidogenesis in mice luteinized follicle cells. Reproduction 2020; 160:955-967. [PMID: 33112769 PMCID: PMC7707808 DOI: 10.1530/rep-20-0340] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/17/2020] [Indexed: 12/29/2022]
Abstract
Luteinization is the event of corpus luteum formation, a way of follicle cells transformation and a process of steroidogenesis alteration. As the core clock gene, Bmal1 was involved in the regulation of ovulation process and luteal function afterwards. Till now, the underlying roles of luteinization played by Bmal1 remain unknown. To explore the unique role of Bmal1 in luteal steroidogenesis and its underlying pathway, we investigated the luteal hormone synthesis profile in Bmal1 knockout female mice. We found that luteal hormone synthesis was notably impaired, and phosphorylation of PI3K/NfκB pathway was significantly activated. Then, the results were verified in in vitro cultured cells, including isolated Bmal1 interference granulosa cells (GCs) and theca cells (TCs), respectively. Hormones levels of supernatant culture media and mRNA expressions of steroidogenesis-associated genes (star, Hsd3β2, cyp19a1 in GCs, Lhcgr, star, Hsd3β2, cyp17a1 in TCs) were mutually decreased, while the phosphorylation of PI3K/NfκB was promoted during in vitro luteinization. After PI3K specific-inhibitor LY294002 intervention, mRNA expressions of Lhcgr and Hsd3β2 were partially rescued in Bmal1 interference TCs, together with significantly increased androstenedione and T synthesis. Further exploration in TCs demonstrated BMAL1 interacted directly but negatively with NfκB p65 (RelA), a subunit which was supposed as a mediator in Bmal1-governed PI3K signaling regulation. Taken together, we verified the novel role of Bmal1 in luteal steroidogenesis, achieving by negative interplay with RelA-mediated PI3K/NfκB pathway.
Collapse
Affiliation(s)
- Yizi Wang
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Minghui Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jian Xu
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou, China
- Reproductive Medicine Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xinyan Liu
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuwei Duan
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Canquan Zhou
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanwen Xu
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
21
|
Yao Y, Feng Q, Shen J. Myosin light chain kinase regulates intestinal permeability of mucosal homeostasis in Crohn's disease. Expert Rev Clin Immunol 2020; 16:1127-1141. [PMID: 33183108 DOI: 10.1080/1744666x.2021.1850269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Researchers have investigated the potential role of intestinal permeability in Crohn's disease pathogenesis. Intestinal permeability is usually mediated by cytoskeleton and intercellular junctions. The myosin light chain kinase (MLCK) is an enzyme that activates the myosin light chain to exert its function related to cytoskeleton contraction and tight junction regulation. The correlation between MLCK and Crohn's disease pathogenesis has been consistently proven. Areas covered: This study aims to expand the understanding of the regulation and function of MLCK in Crohn's disease. An extensive literature search in the MEDLINE database (via PubMed) has been performed up to Oct. 2020. The roles of MLCK in tight junction activation, intestinal permeability enhancement, and cell signal regulation are comprehensively discussed. Expert opinion: Targeting the MLCK-related pathways such as TNF-α in CD treatment has been put into clinical use. More accurate targeting such as MLCK and TNFR2 has been proposed to reduce side effects. MLCK may also have the potential to become biomarkers in fields like CD activity. With the application of cutting age research methods and tools, the MLCK research could be accelerated.
Collapse
Affiliation(s)
- Yiran Yao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center; Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University , Shanghai, China
| | - Qi Feng
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center; Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University , Shanghai, China
| |
Collapse
|
22
|
Imatinib exhibit synergistic pleiotropy in the prevention of colorectal cancer by suppressing proinflammatory, cell survival and angiogenic signaling. Cell Signal 2020; 76:109803. [PMID: 33022360 DOI: 10.1016/j.cellsig.2020.109803] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 12/24/2022]
Abstract
Recent global incidences and mortality rates have placed colorectal cancer (CRC) at third and second positions, respectively, among both sexes of all ages. Resistance during chemotherapy is a big problem in the treatment and disease-free survival of CRC patients. Discovery of new anticancer drug(s) is a time taking process and therefore, invites studies for repurposing the known therapeutics. The present study was conceived to analyze the anticancer role of Imatinib in experimental CRC at early stages. Different experimental procedures e.g. tumor incidences or histoarchitectural changes, gene and protein expression analysis, estimations of intracellular calcium, ROS, mitochondrial membrane potential, apoptotic index and molecular docking was performed to support the hypothesis. It was observed that Imatinib could function as an immunomodulator by breaking the feed-back loop between the proinflammatory cytokines (IL-1β and TNF-α) and transcription factors (NF-κB, Jak3/Stat3) knowingly involved in increased cell proliferation during tumorigenesis via activating different intracellular signaling. Also, Imatinib could independently deregulate the other cell survival and proliferation signaling e.g. PI3-K/Akt/mTOR, Wnt/β-catenin and MAPK. Proinflammatory cytokines orchestrated intracellular signaling also involve angiogenic factors to be upregulated during CRC which were also seemed to be independently suppressed by Imatinib. Restoration of physiological apoptosis by increasing the release of intracellular calcium to generate ROS thereby reducing the mitochondrial membrane potential for the release of cytochrome c and activation of caspase-3 was also reported with Imatinib administration. Thus, it may be suggested that Imatinib show synergistic pleiotropy in suppressing the interlinked tumorigenic signaling pathways independently.
Collapse
|
23
|
Autophagy regulates trophoblast invasion by targeting NF-κB activity. Sci Rep 2020; 10:14033. [PMID: 32820194 PMCID: PMC7441061 DOI: 10.1038/s41598-020-70959-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
Preeclampsia is one of the most serious complications of pregnancy, affecting 5-10% of parturients worldwide. Recent studies have suggested that autophagy is involved in trophoblast invasion and may be associated with defective placentation underlying preeclampsia. We thus aimed to understand the mechanistic link between autophagy and trophoblast invasion. Using the two most commonly used trophoblast cell lines, JEG-3 and HTR-8/SVneo, we inhibited autophagy by ATG5 and beclin-1 shRNA. Conversion of LC3-II was evaluated in ATG5 and beclin-1 knock-down cells in the presence of the lysosomal protease inhibitors E-64d and pepstatin A, to detect the efficiency of autophagy inhibition. Upon autophagy inhibition, we measured cell invasion, activity of NF-κB and related signaling pathways, MMP-2, MMP-9, sFlt-1, and TNF-α levels. Autophagy inhibition increased the invasiveness of these trophoblastic cell lines and increased Akt and NF-κB activity as well as p65 expression. Of note, an NF-κB inhibitor significantly attenuated the trophoblast invasion induced by autophagy inhibition. Autophagy inhibition was also associated with increased MMP-2 and MMP-9 levels and decreased the production of sFlt-1 and TNF-α. Collectively, our results indicate that autophagy regulates trophoblast invasiveness in which the NF-κB pathway and MMP-2, MMP-9, sFlt-1 and TNF-α levels are affected.
Collapse
|
24
|
Doukas SG, Doukas PG, Sasaki CT, Vageli D. The in vivo preventive and therapeutic properties of curcumin in bile reflux-related oncogenesis of the hypopharynx. J Cell Mol Med 2020; 24:10311-10321. [PMID: 32691972 PMCID: PMC7521262 DOI: 10.1111/jcmm.15640] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/21/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
Bile at strongly acidic pH exerts a carcinogenic effect on the hypopharynx, based upon recent pre‐clinical studies that support its role as an independent risk factor. We recently demonstrated in vitro that curcumin can prevent oncogenic profile of bile in human hypopharyngeal cells, by inhibiting NF‐κB. We hypothesize that topically applied curcumin to the hypopharynx can similarly block early oncogenic molecular events of bile, by inhibiting NF‐κB and consequently altering the expression of genes with oncogenic function. Using Mus musculus (C57Bl/6J), we topically applied curcumin (250 μmol/L; three times per day; 10 days) to the hypopharynx, 15 minutes before, 15 minutes after or in combination with bile acids (pH 3.0). Immunohistochemical analysis and qPCR revealed that topically applied curcumin either before, after or in combination with acidic bile exposure significantly suppressed its induced NF‐κB activation in regenerating epithelial cells, and overexpression of Rela, Bcl2, Egfr, Stat3, Wnt5a, Tnf, Il6, Ptgs2. Akt1 was particularly inhibited by curcumin when applied simultaneously with bile. We provide novel evidence into the preventive and therapeutic properties of topically applied curcumin in acidic bile‐induced early oncogenic molecular events in hypopharyngeal mucosa, by inhibiting NF‐κB, and shaping future translational development of effective targeted therapies using topical non‐pharmacologic inhibitors of NF‐κB.
Collapse
Affiliation(s)
- Sotirios G Doukas
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Panagiotis G Doukas
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Clarence T Sasaki
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Dimitra Vageli
- The Yale Larynx Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
25
|
Wang X, Liu Y, Zhang S, Ouyang X, Wang Y, Jiang Y, An N. Crosstalk between Akt and NF-κB pathway mediates inhibitory effect of gas6 on monocytes-endothelial cells interactions stimulated by P. gingivalis-LPS. J Cell Mol Med 2020; 24:7979-7990. [PMID: 32462812 PMCID: PMC7348146 DOI: 10.1111/jcmm.15430] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/01/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023] Open
Abstract
Correlation between periodontitis and atherosclerosis is well established, and the inherent mechanisms responsible for this relationship remain unclear. The biological function of growth arrest‐specific 6 (gas6) has been discovered in both atherosclerosis and inflammation. Inhibitory effects of gas6 on the expression of inflammatory factors in human umbilical vein endothelial cells (HUVECs) stimulated by Porphyromonas gingivalis lipopolysaccharide (P. gingivalis‐LPS) were reported in our previous research. Herein, the effects of gas6 on monocytes‐endothelial cells interactions in vitro and their probable mechanisms were further investigated. Gas6 protein in HUVECs was knocked down with siRNA or overexpressed with plasmids. Transwell inserts and co‐culturing system were introduced to observe chemotaxis and adhering affinity between monocytes and endothelial cells in vitro. Expression of gas6 was decreased in inflammatory periodontal tissues and HUVECs challenged with P. gingivalis‐LPS. The inhibitory effect of gas6 on chemotaxis and adhesion affinity between monocytes and endothelial cells was observed, and gas6 promoted Akt phosphorylation and inhibited NF‐κB phosphorylation. To our best knowledge, we are first to report that gas6 inhibit monocytes‐endothelial cells interactions in vitro induced by P. gingivalis‐LPS via Akt/NF‐κB pathway. Additionally, inflammation‐mediated inhibition of gas6 expression is through LncRNA GAS6‐AS2, rather than GAS6‐AS1, which is also newly reported.
Collapse
Affiliation(s)
- Xuekui Wang
- Department of General Dentistry II, Peking University School and Hospital of Stomatology, Beijing, China.,Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yingjun Liu
- Department of General Dentistry II, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Shengnan Zhang
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiangying Ouyang
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yuguang Wang
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yong Jiang
- Department of General Dentistry II, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Na An
- Department of General Dentistry II, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
26
|
Hu Y, Wei M, Niu Q, Ma R, Li Y, Wang X, Feng G, Li S, Pang L. Grape seed proanthocyanidin extract alleviates arsenic-induced lung damage through NF-κB signaling. Exp Biol Med (Maywood) 2020; 244:213-226. [PMID: 30869553 DOI: 10.1177/1535370219829881] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
IMPACT STATEMENT Arsenic-induced respiratory inflammatory damage is an important occupational hazard in many areas of the world, particularly in underdeveloped and developing countries. Effective treatments are lacking and expensive. Therefore, the aim of the study was to examine the anti-inflammatory effects of proanthocyanidin (PC) and the molecular mechanisms in vivo and in vitro. The present study showed that PC extracted from grape seed could attenuate the lung damage in a mouse model of arsenic poisoning. The effects were observed at the level of lung histology and inflammasome expression. This study suggests that a natural compound is effective in mitigating the toxic effects of arsenic in the lungs, providing an inexpensive and more readily accessible method for treating arsenic exposure in some parts of the world.
Collapse
Affiliation(s)
- Yunhua Hu
- 1 Department of Public Health, Shihezi University School of Medicine, Shihezi 832003, China
| | - Meng Wei
- 1 Department of Public Health, Shihezi University School of Medicine, Shihezi 832003, China.,2 Department of Outpatient, The First Affiliated Hospital, Xinjiang Medical University, Xinjiang 830054, China
| | - Qiang Niu
- 1 Department of Public Health, Shihezi University School of Medicine, Shihezi 832003, China
| | - Rulin Ma
- 1 Department of Public Health, Shihezi University School of Medicine, Shihezi 832003, China
| | - Yu Li
- 1 Department of Public Health, Shihezi University School of Medicine, Shihezi 832003, China
| | - Xianhua Wang
- 3 Department of Quality Control of Changji Hui Autonomous Prefecture Center for Disease Control and Prevention in the Xinjiang Uygur Autonomous Region, Changji, Xinjiang 831100, China
| | - Gangling Feng
- 1 Department of Public Health, Shihezi University School of Medicine, Shihezi 832003, China
| | - Shugang Li
- 1 Department of Public Health, Shihezi University School of Medicine, Shihezi 832003, China
| | - Lijuan Pang
- 1 Department of Public Health, Shihezi University School of Medicine, Shihezi 832003, China
| |
Collapse
|
27
|
Kiang JG, Smith JT, Cannon G, Anderson MN, Ho C, Zhai M, Cui W, Xiao M. Ghrelin, a novel therapy, corrects cytokine and NF-κB-AKT-MAPK network and mitigates intestinal injury induced by combined radiation and skin-wound trauma. Cell Biosci 2020; 10:63. [PMID: 32426105 PMCID: PMC7216502 DOI: 10.1186/s13578-020-00425-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 05/04/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Compared to radiation injury alone (RI), radiation injury combined wound (CI) further enhances acute radiation syndrome and subsequently mortality. We previously reported that therapy with Ghrelin, the 28-amino-acid-peptide secreted from the stomach, significantly increased 30-day survival and mitigated hematopoietic death by enhancing and sustaining granulocyte-colony stimulating factor (G-CSF) and keratinocyte chemoattractant (KC) in the blood and bone marrow; increasing circulating white blood cell depletion; inhibiting splenocytopenia; and accelerating skin-wound healing on day 30 after CI. Herein, we aimed to study the efficacy of Ghrelin on intestinal injury at early time points after CI. METHODS B6D2F1/J female mice were exposed to 60Co-γ-photon radiation (9.5 Gy, 0.4 Gy/min, bilateral), followed by 15% total-body-surface-area skin wounds. Several endpoints were measured: at 4-5 h and on days 1, 3, 7, and 15. RESULTS Ghrelin therapy mitigated CI-induced increases in IL-1β, IL-6, IL-17A, IL-18, KC, and TNF-α in serum but sustained G-CSF, KC and MIP-1α increases in ileum. Histological analysis of ileum on day 15 showed that Ghrelin treatment mitigated ileum injury by increasing villus height, crypt depth and counts, as well as decreasing villus width and mucosal injury score. Ghrelin therapy increased AKT activation and ERK activation; suppressed JNK activation and caspase-3 activation in ileum; and reduced NF-κB, iNOS, BAX and Bcl-2 in ileum. This therapy recovered the tight junction protein and mitigated bacterial translocation and lipopolysaccharides levels. The results suggest that the capacity of Ghrelin therapy to reduce CI-induced ileum injury is mediated by a balanced NF-κB-AKT-MAPK network that leads to homeostasis of pro-inflammatory and anti-inflammatory cytokines. CONCLUSIONS Our novel results are the first to suggest that Ghrelin therapy effectively decreases intestinal injury after CI.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, MD 20814 USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services, University of the Health Sciences, Bethesda, MD 20814 USA
- Department of Medicine, Uniformed Services, University of the Health Sciences, Bethesda, MD 20814 USA
| | - Joan T. Smith
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, MD 20814 USA
| | - Georgetta Cannon
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, MD 20814 USA
| | - Marsha N. Anderson
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, MD 20814 USA
| | - Connie Ho
- Department of Biochemistry, University of California, Berkeley, CA 94720 USA
| | - Min Zhai
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, MD 20814 USA
| | - Wanchang Cui
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, MD 20814 USA
| | - Mang Xiao
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, MD 20814 USA
| |
Collapse
|
28
|
Vallée A, Lecarpentier Y. Curcumin and Endometriosis. Int J Mol Sci 2020; 21:E2440. [PMID: 32244563 PMCID: PMC7177778 DOI: 10.3390/ijms21072440] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 12/14/2022] Open
Abstract
Endometriosis is one of the main common gynecological disorders, which is characterized by the presence of glands and stroma outside the uterine cavity. Some findings have highlighted the main role of inflammation in endometriosis by acting on proliferation, apoptosis and angiogenesis. Oxidative stress, an imbalance between reactive oxygen species and antioxidants, could have a key role in the initiation and progression of endometriosis by resulting in inflammatory responses in the peritoneal cavity. Nevertheless, the mechanisms underlying this disease are still unclear and therapies are not currently efficient. Curcumin is a major anti-inflammatory agent. Several findings have highlighted the anti-oxidant, anti-inflammatory and anti-angiogenic properties of curcumin. The purpose of this review is to summarize the potential action of curcumin in endometriosis by acting on inflammation, oxidative stress, invasion and adhesion, apoptosis and angiogenesis.
Collapse
Affiliation(s)
- Alexandre Vallée
- Diagnosis and Therapeutic Center, Hypertension and Cardiovascular Prevention Unit, Hôtel-Dieu Hospital, AP-HP, Paris-Descartes University, 75004 Paris, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien (GHEF), 77100 Meaux, France;
| |
Collapse
|
29
|
Kaur S, Kumar A, Thakur S, Kumar K, Sharma R, Sharma A, Singh P, Sharma U, Kumar S, Landi M, Brestič M, Kaur S. Antioxidant, Antiproliferative and Apoptosis-Inducing Efficacy of Fractions from Cassia fistula L. Leaves. Antioxidants (Basel) 2020; 9:E173. [PMID: 32093300 PMCID: PMC7070616 DOI: 10.3390/antiox9020173] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/15/2020] [Accepted: 02/17/2020] [Indexed: 02/07/2023] Open
Abstract
: Cassia fistula L. is a highly admirable traditional medicinal plant used for the treatment of various diseases and disorders. The present study was performed to divulge the antioxidant, antiproliferative, and apoptosis-inducing efficacy of fractions from C. fistula leaves. The hexane (CaLH fraction), chloroform (CaLC fraction), ethyl acetate (CaLE fraction), n-butanol (CaLB fraction), and aqueous (CaLA fraction) were sequentially fractionated from 80% methanolic (CaLM extract) of C. fistula leaves. The CaLE fraction was fractionated using column chromatography to yield a pure compound, which was characterized as Epiafzelechin (CFL1) based on 1H, 13C, and DEPT135 NMR. Among these fractions, CaLE and isolated CFL1 fractions exhibited an effective antioxidant potential in Ferric ion reducing power, (2,2'-azino-bis (3-ethylbenzothiazoline -6-sulfonic acid)) cation radical scavenging, and nitric oxide radical scavenging assays. Epiafzelechin was investigated for its antiproliferative effects against MG-63 (osteosarcoma), IMR-32 (neuroblastoma), and PC-3 (prostate adenocarcinoma), and was found to inhibit cell proliferation with a GI50 value of 8.73, 9.15, and 11.8 μM respectively. MG-63 cells underwent apoptotic cell death on treatment with Epiafzelechin as the cells showed the formation of apoptotic bodies, enhanced reactive oxygen species (ROS) generation, mitochondrial membrane depolarization along with an increase in early apoptotic cell population analyzed using Annexin V-FITC/PI double staining assay. Cells showed cell cycle arrest at the G0/G1 phase accompanied by a downregulation in the expression levels of p-Akt (Protein kinase B), p-GSK-3β (Glycogen synthase kinase-3 beta), and Bcl-xl (B-cell lymphoma-extra large) proteins. RT-PCR (Real time-polymerase chain reaction) analysis revealed downregulation in the gene expression level of β-catenin and CDK2 (cyclin-dependent kinases-2) while it upregulated the expression level of caspase-8 and p53 genes in MG-63 cells.
Collapse
Affiliation(s)
- Sandeep Kaur
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar 143005, India; (S.K.); (A.K.); (A.S.)
| | - Ajay Kumar
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar 143005, India; (S.K.); (A.K.); (A.S.)
| | - Sharad Thakur
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar 143005, India;
| | - Kapil Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar 143005, India; (K.K.); (P.S.); (S.K.)
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab 144411, India
| | - Ritika Sharma
- Natural Product Chemistry and Process Development division, CSIR-IHBT, Palampur 176061, India; (R.S.); (U.S.)
| | - Anket Sharma
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar 143005, India; (S.K.); (A.K.); (A.S.)
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, China
| | - Prabhpreet Singh
- Department of Chemistry, Guru Nanak Dev University, Amritsar 143005, India; (K.K.); (P.S.); (S.K.)
| | - Upendra Sharma
- Natural Product Chemistry and Process Development division, CSIR-IHBT, Palampur 176061, India; (R.S.); (U.S.)
| | - Subodh Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar 143005, India; (K.K.); (P.S.); (S.K.)
| | - Marco Landi
- Department of Agriculture, Food and Environment, University of Pisa, 56124 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
- CIRSEC, Centre for Climatic Change Impact, University of Pisa, 56124 Pisa, Italy
| | - Marián Brestič
- Department of Plant Physiology, Faculty of Agrobiology and Food Resources, Slovak University of Agriculture, 94976 Nitra, Slovakia;
- Department of Botany and Plant Physiology, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, 16500 Prague, Czech Republic
| | - Satwinderjeet Kaur
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar 143005, India; (S.K.); (A.K.); (A.S.)
| |
Collapse
|
30
|
MicroRNA-31 regulating apoptosis by mediating the phosphatidylinositol-3 kinase/protein kinase B signaling pathway in treatment of spinal cord injury. Brain Dev 2019; 41:649-661. [PMID: 31036380 DOI: 10.1016/j.braindev.2019.04.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/01/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022]
Abstract
Apoptosis is a highly conservative energy demand program for non-inflammatory cell death, which is extremely significant in normal physiology and disease. There are many techniques used for studying apoptosis. MicroRNA (miRNA) is closely related to cell apoptosis, and especially microRNA-31 (miR-31) is involved in apoptosis by regulating a large number of target genes and signaling pathways. In many neurological diseases, cell apoptosis or programmed cell death plays an important role in the reduction of cell number, including the reduction of neurons in spinal cord injuries. In recent years, the phosphoinositol 3-kinase/AKT (PI3K/AKT) signal pathway, as a signal pathway involved in a variety of cell functions, has been studied in spinal cord injury diseases. The PI3K/AKT pathway directly or indirectly affects whether apoptosis occurs in a cell, thereby affecting a significant intracellular event sequence. This paper reviewed the interactions of miR-31 target sites in the PI3K/AKT signaling pathway, and explored new ways to prevent and treat spinal cord injury by regulating the effect of miR-31 on apoptosis.
Collapse
|
31
|
Kaur H, Moreau R. Role of mTORC1 in intestinal epithelial repair and tumorigenesis. Cell Mol Life Sci 2019; 76:2525-2546. [PMID: 30944973 PMCID: PMC11105546 DOI: 10.1007/s00018-019-03085-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/08/2019] [Accepted: 03/25/2019] [Indexed: 12/15/2022]
Abstract
mTORC1 signaling is the prototypical pathway regulating protein synthesis and cell proliferation. mTORC1 is active in stem cells located at the base of intestinal crypts but silenced as transit-amplifying cells differentiate into enterocytes or secretory cells along the epithelium. After an insult or injury, self-limiting and controlled activation of mTORC1 is critical for the renewal and repair of intestinal epithelium. mTORC1 promotes epithelial cell renewal by driving cryptic stem cell division, and epithelial cell repair by supporting the dedifferentiation and proliferation of enterocytes or secretory cells. Under repeated insult or injury, mTORC1 becomes constitutively active, triggering an irreversible return to stemness, cell division, proliferation, and inflammation among dedifferentiated epithelial cells. Epithelium-derived cytokines promulgate inflammation within the lamina propria, which in turn releases inflammatory factors that act back on the epithelium where undamaged intestinal epithelial cells participate in the pervading state of inflammation and become susceptible to tumorigenesis.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
32
|
Zhang J, Zhang Y, Wang J, Zhang S, Zhao Y, Ren H, Chu Y, Feng L, Wang C. Protein kinase D3 promotes gastric cancer development through p65/6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 activation of glycolysis. Exp Cell Res 2019; 380:188-197. [PMID: 31026442 DOI: 10.1016/j.yexcr.2019.04.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 11/16/2022]
Abstract
Although serine/threonine-protein kinases are found to participate in a wide range of cancer progression, the involvement of protein kinase D3 (PRKD3) in gastric cancer has not been explored. Here, we investigated the role of PRKD3 in gastric cancer (GC) and its potential mechanisms. PRKD3 was over-expressed in gastric cancer tissues and cells. In vitro, PRKD3 ectopic expression accelerated the proliferation and growth of GES-1, SGC7901 and MKN-28 cells. By contrast, PRKD3 knockdown suppressed the proliferation of SGC7901 and MKN-28 GC cells. In vivo, xenograted tumorigenesis was blunted by PRKD3 silencing. Mechanistically, PRKD3 up-regulated 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) and activated glycolysis as shown by increased glucose consumption and lactate production. Knockdown of PFKFB3 suppressed the glycolysis in gastric cancer cells with highly expressed PRKD3 but not in PRKD3 silenced cells. PRKD3 over-expression induced phosphorylation of p65 at serine 536 was critical for the up-regulation of glycolytic enzyme PFKFB3. Furthermore, PRKD and PFKFB3 inhibitor suppressed the viability of GC cells. Our results suggest that targeting PRKD3/p65/PFKFB3 cascade maybe a promising therapeutic strategy for gastric cancer.
Collapse
Affiliation(s)
- Jianwei Zhang
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yajing Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiaqi Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuisheng Zhang
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yajie Zhao
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hu Ren
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yunmian Chu
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Chengfeng Wang
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
33
|
Samimi M, Pourhanifeh MH, Mehdizadehkashi A, Eftekhar T, Asemi Z. The role of inflammation, oxidative stress, angiogenesis, and apoptosis in the pathophysiology of endometriosis: Basic science and new insights based on gene expression. J Cell Physiol 2019; 234:19384-19392. [PMID: 31004368 DOI: 10.1002/jcp.28666] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 01/24/2023]
Abstract
Endometriosis is a frequent and chronic illness in young women which could be defined by the existence of endometrial stroma and glands outside of the normal site of the lining of the uterus. It has painful symptoms. The advanced stage of endometriosis may lead to gynecological malignancies, such as ovarian cancer, and other complications, including infertility. However, its exact physiopathology is not well known. Recent studies have shown the possible roles of inflammation along with oxidative stress. Additionally, angiogenesis and apoptosis dysregulation contribute to endometriosis pathophysiology. Therapeutic strategies and continuing attempts, to conquer endometriosis should be done regarding molecular signaling pathways. Thus, the present review summarizes current studies and focuses on molecular mechanisms.
Collapse
Affiliation(s)
- Mansooreh Samimi
- Endometriosis Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Tahereh Eftekhar
- Reproductive Health Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
34
|
Role of ANTXR1 in the regulation of RANKL-induced osteoclast differentiation and function. Biochem Biophys Res Commun 2019; 510:296-302. [PMID: 30686531 DOI: 10.1016/j.bbrc.2019.01.094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/19/2019] [Indexed: 11/21/2022]
Abstract
Anthrax toxin receptor 1 (ANTXR1) is a transmembrane protein with an extracellular domain which is deeply associated with the process of bone formation and plays an important role in angiogenesis. However, there have been no reports investigating the effects of ANTXR1 on bone metabolism mediated by the two types of bone cells, osteoclasts, and osteoblasts. The aim of this study is to reveal the role of ANTXR1 in the differentiation and function of osteoclasts and osteoblasts. We found that ANTXR1 positively regulated the receptor activator of nuclear factor kappa B ligand (RANKL)-induced osteoclast differentiation and bone resorption with no effects on osteoblast differentiation by performing gain- and loss-of-function studies. During ANTXR1-mediated regulation of osteoclastogenesis, phosphorylation of early signal transducers such as c-Jun N-terminal kinase (JNK), Akt, inhibitor of kappa B (IκB), and phospholipase C gamma 2 (PLCγ2) was affected, which in turn altered the mRNA and protein levels of c-Fos and nuclear factor of activated T cells cytoplasmic 1 (NFATc1). In addition, genetic manipulation of ANTXR1 in bone marrow macrophages (BMMs) modulated the capillary-like tube formation in HUVECs via secretion of two angiogenic factors, matrix metalloproteinase-9 (MMP-9) and vascular endothelial growth factor-A (VEGF-A). These results elucidated the importance of ANTXR1 in osteoclast differentiation and functional activity, as well as, osteoclast-mediated angiogenesis of endothelial cells. Taken together, we propose that ANTXR1 might be a promising candidate for gene therapy for bone metabolic diseases and further, might potentially serve as an important biomarker in the field of bone metastasis associated with vascularization.
Collapse
|
35
|
Möser CV, Möller M, Fleck SC, Thomas D, Geisslinger G, Niederberger E. Inhibition of the protein kinase IKKepsilon attenuates neuropathic pain in mice. Neuropharmacology 2018; 146:198-211. [PMID: 30528326 DOI: 10.1016/j.neuropharm.2018.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 11/09/2018] [Accepted: 12/05/2018] [Indexed: 12/30/2022]
Abstract
Inhibitor-kappaB kinase epsilon (IKKε, Ikbke) constitutes an NF-κB activating kinase with high homology to the classical I-κB kinase subunits, IKKα and IKKβ. It is expressed in nociceptive neurons in the spinal cord and in dorsal root ganglia (DRG) and involved in inflammatory nociception. Under inflammatory conditions, IKKε deficient mice show significantly less nociceptive behavior in comparison to wild type mice associated with reduced activation of NF-κB and attenuated NF-κB-dependent gene expression. The role of IKKε in neuropathic pain has not been investigated so far. We applied the spared nerve injury (SNI) model of neuropathic pain in mice and found an increased expression of IKKε in the spinal cord, the DRGs and the sciatic nerve after induction of neuropathy. Genetic depletion of IKKε or pharmacological inhibition by amlexanox led to a significant reduction of mechanical hyperalgesia and cold allodynia in comparison to control mice. Transcription factor ELISA indicated that the effects are mediated by reduced activation of NF-κB. Furthermore, immunofluorescence staining, qPCR and Western Blot analyses revealed that the decreased pain-like behavior was associated with a reduced activation of microglia, diminished expression of c-fos as well as a decreased activation of MAP-Kinases. In summary, we conclude that IKKε modulates mechanisms of neuropathic pain by activating NF-κB. The administration of IKKε inhibitors might therefore constitute a new and promising approach for the therapy of neuropathic pain.
Collapse
Affiliation(s)
- Christine V Möser
- pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt am Main, Germany.
| | - Moritz Möller
- pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt am Main, Germany.
| | - Stefanie C Fleck
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group for Translational Medicine & Pharmacology TMP, 60596, Frankfurt am Main, Germany.
| | - Dominique Thomas
- pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt am Main, Germany.
| | - Gerd Geisslinger
- pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt am Main, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group for Translational Medicine & Pharmacology TMP, 60596, Frankfurt am Main, Germany.
| | - Ellen Niederberger
- pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
36
|
Asghari S, Valizadeh Dizajeykan A, Ahmadi M, Barzegari A, Rikhtegar R, Dolati S, Danaii S, Abdollahi‐Fard S, Nouri M, Mahdipour M, Yousefi M. Evaluation of ovarian cancer risk in granulosa cells treated with steroid‐depleted endometriosis serum: Role of NF‐κB/RelA and AKT. J Cell Physiol 2018; 234:12011-12018. [DOI: 10.1002/jcp.27862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/13/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Samira Asghari
- Students’ Research Committee, Tabriz University of Medical Sciences Tabriz Iran
| | | | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Abolfazl Barzegari
- Research Center of Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences Tabriz Iran
| | - Reza Rikhtegar
- Aging Research Institute, Tabriz University of Medical Sciences Tabriz Iran
| | - Sanam Dolati
- Aging Research Institute, Tabriz University of Medical Sciences Tabriz Iran
| | - Shahla Danaii
- Department of Gynecology Eastern Azerbaijan ACECR ART Center, Eastern Azerbaijan Branch of ACECR Tabriz Iran
| | - Sedigheh Abdollahi‐Fard
- Department of Gynecology Eastern Azerbaijan ACECR ART Center, Eastern Azerbaijan Branch of ACECR Tabriz Iran
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Reproductive Biology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Reproductive Biology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
37
|
Bale S, Pulivendala G, Godugu C. Withaferin A attenuates bleomycin-induced scleroderma by targeting FoxO3a and NF-κβ signaling: Connecting fibrosis and inflammation. Biofactors 2018; 44:507-517. [PMID: 30367690 DOI: 10.1002/biof.1446] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 07/24/2018] [Indexed: 12/16/2022]
Abstract
Scleroderma is an inflammatory autoimmune disease which begins with inflammation due to tissue injury and advances to progressive accumulation of extracellular matrix resulting in scarring and hardening of the skin. Inflammation is a salutary response to tissue injury caused by varied factors. While inflammation is required for systematic wound healing, dysregulated chronic inflammation often leads to tissue scarring. Prominent role of inflammation in pathology and physiology makes it a double edge sword. The objective of this study was to investigate the role of Withaferin A (WFA), a steroidal lactone from Withania somnifera in a 28-day murine model of bleomycin-induced experimental scleroderma. Withaferin A was administered at two doses 2 and 4 mg/kg intraperitoneally for 28 days. At the time of study termination, we observed significant reduction in dorsal skin thickness. Our results indicate that WFA was able to sufficiently suppress pro-inflammatory phase of fibrosis, TGF-β/Smad signaling and also significantly repressed fibroblast conversion to myofibroblasts. Additionally, our study also demonstrated that WFA modulates FoxO3a-Akt-dependent NF-κβ/IKK-mediated inflammatory cascade, which is a prime signaling pathway in fibrogenesis. The findings of this study are persuasive of WFA as an antifibrotic agent with promising therapeutic effects in scleroderma. © 2018 BioFactors, 44(6):507-517, 2018.
Collapse
Affiliation(s)
- Swarna Bale
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Gauthami Pulivendala
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| |
Collapse
|
38
|
Verma AK, Waghmare TS, Jachak GR, Philkhana SC, Reddy DS, Basu A. Nitrosporeusine analogue ameliorates Chandipura virus induced inflammatory response in CNS via NFκb inactivation in microglia. PLoS Negl Trop Dis 2018; 12:e0006648. [PMID: 30001342 PMCID: PMC6063446 DOI: 10.1371/journal.pntd.0006648] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 07/27/2018] [Accepted: 06/28/2018] [Indexed: 12/30/2022] Open
Abstract
Chandipura Virus (CHPV), a negative-stranded RNA virus belonging to the Rhabdoviridae family, has been previously reported to bring neuronal apoptosis by activating several factors leading to neurodegeneration. Following virus infection of the central nervous system, microglia, the ontogenetic and functional equivalents of macrophages in somatic tissues gets activated and starts secreting chemokines, thereby recruiting peripheral leukocytes into the brain parenchyma. In the present study, we have systemically examined the effect of CHPV on microglia and the activation of cellular signalling pathways leading to chemokine expression upon CHPV infection. Protein and mRNA expression profiles of chemokine genes revealed that CHPV infection strongly induces the expression of CXC chemokine ligand 10 (CXCL10) and CC chemokine ligand 5 (CCL5) in microglia. CHPV infection triggered the activation of signalling pathways mediated by mitogen-activated protein kinases, including p38, JNK 1 and 2, and nuclear factor κB (NF-kappaB). CHPV-induced expression of CXCL10 and CCL5 was achieved by the activation of p38 and NF-kappaB pathways. Considering the important role of inflammation in neurodegeneration, we have targeted NF-kappaB using a newly synthesised natural product nitrosporeusine analogue and showed incapability of microglial supernatant of inducing apoptosis in neurons after treatment.
Collapse
Affiliation(s)
| | - Trushnal S. Waghmare
- National Brain Research Centre, Manesar, Haryana, India
- National Institute of Virology, Pune, India
| | | | | | | | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, India
- * E-mail:
| |
Collapse
|
39
|
Crasto JA, Fourman MS, Morales-Restrepo A, Mahjoub A, Mandell JB, Ramnath K, Tebbets JC, Watters RJ, Weiss KR. Disulfiram reduces metastatic osteosarcoma tumor burden in an immunocompetent Balb/c or-thotopic mouse model. Oncotarget 2018; 9:30163-30172. [PMID: 30046395 PMCID: PMC6059028 DOI: 10.18632/oncotarget.25733] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/14/2018] [Indexed: 12/16/2022] Open
Abstract
Introduction The overall survival rate of patients with osteosarcoma (OS) and pulmonary metastases has remained stagnant at 15–30% for several decades. Disulfiram (DSF) is an FDA-approved aldehyde dehydrogenase inhibitor that reduces the metastatic phenotype of OS cells in vitro. Here we evaluate its in vivo efficacy, as compared to doxorubicin chemotherapy, in a previously-validated orthotopic model of metastatic OS. Results All treatment groups displayed a significantly reduced quantitative OS metastatic burden compared with controls. The metastatic burden of Lo DSF-treated animals was equivalent to the DXR group. Ninety-five percent of control animals displayed evidence of metastatic disease, which was significantly greater than all treatment groups. Discussion Disulfiram treatment resulted in a reduced burden of OS metastatic disease compared with controls. This was statistically-equivalent to doxorubicin. No additive effect was observed between these two therapies. Materials and Methods One-hundred twenty immunocompetent Balb/c mice received proximal tibia paraphyseal injections of 5 × 105 K7M2 murine OS cells. Therapy began three weeks after injection: saline (control), low-dose disulfiram (Lo DSF), high-dose disulfiram (Hi DSF), doxorubicin (DXR), Lo DSF + DXR, and Hi DSF + DXR. Transfemoral amputations were performed at 4 weeks. Quantitative metastatic tumor burden was measured using near-infrared indocyanine green (ICG) angiography.
Collapse
Affiliation(s)
- Jared Anthony Crasto
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mitchell Stephen Fourman
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alejandro Morales-Restrepo
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adel Mahjoub
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan Brendan Mandell
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kavita Ramnath
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jessica C Tebbets
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rebecca J Watters
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, PA, USA
| | - Kurt Richard Weiss
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Departments of Anatomic Pathology and General Surgical Oncology, University of Pittsburgh, PA, USA
| |
Collapse
|
40
|
Klobučar M, Grbčić P, Pavelić SK, Jonjić N, Visentin S, Sedić M. Acid ceramidase inhibition sensitizes human colon cancer cells to oxaliplatin through downregulation of transglutaminase 2 and β1 integrin/FAK-mediated signalling. Biochem Biophys Res Commun 2018; 503:843-848. [PMID: 29920241 DOI: 10.1016/j.bbrc.2018.06.085] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 06/16/2018] [Indexed: 01/14/2023]
Abstract
Acid ceramidase (ASAH1) has been implicated in the progression and chemoresistance in different cancers. Its role in colon cancer biology and response to standard chemotherapy has been poorly addressed so far. Here, we have investigated ASAH1 expression at the protein level in human colon cancer cell lines and tissues from colon cancer patients, and have examined in vitro the possible link between ASAH1 expression and functional activity of p53 protein whose inactivation is associated with the progression from adenoma to malignant tumour in colon cancer. Finally, we have explored the role of ASAH1 in response and resistance mechanisms to oxaliplatin (OXA) in HCT 116 colon cancer cells. We have demonstrated that human colon cancer cells and colorectal adenocarcinoma tissues constitutively express ASAH1, and that its expression is higher in tumour tissues than in normal colonic mucosa. Furthermore, we found an inverse correlation between ASAH1 expression and p53 functional activity. Obtained data revealed that ASAH1 was involved in HCT 116 cell response to OXA and that anti-proliferative, pro-apoptotic, anti-migratory and anti-clonogenic effects of OXA could be significantly increased by combination treatment with ASAH1 inhibitor carmofur. Increased OXA sensitivity was associated with downregulation of signalling involved in acquired resistance to OXA in colon cancer, in particular transglutaminase 2 and β1 integrin/FAK, which resulted in the suppression of NF-κB and Akt. Thus, combination of OXA with ASAH1 inhibitors could be a promising strategy to counter chemoresistance and improve treatment outcome in advanced colon cancer.
Collapse
Affiliation(s)
- Marko Klobučar
- University of Rijeka Department of Biotechnology, Centre for High-Throughput Technologies, Radmile Matejčić 2, 51000 Rijeka, Croatia
| | - Petra Grbčić
- University of Rijeka Department of Biotechnology, Centre for High-Throughput Technologies, Radmile Matejčić 2, 51000 Rijeka, Croatia
| | - Sandra Kraljević Pavelić
- University of Rijeka Department of Biotechnology, Centre for High-Throughput Technologies, Radmile Matejčić 2, 51000 Rijeka, Croatia
| | - Nives Jonjić
- University of Rijeka Faculty of Medicine, Department for General Pathology and Pathologic Anatomy, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Sarah Visentin
- University of Rijeka Department of Biotechnology, Centre for High-Throughput Technologies, Radmile Matejčić 2, 51000 Rijeka, Croatia
| | - Mirela Sedić
- University of Rijeka Department of Biotechnology, Centre for High-Throughput Technologies, Radmile Matejčić 2, 51000 Rijeka, Croatia.
| |
Collapse
|
41
|
Li ZP, Li LF, Zhang QW, Wei W, Liu HB, Bao WR, Ma DL, Leung CH, Bian ZX, Lu AP, Han QB. Akt downstream of NFκB, MAPKs and IRF3 pathway involved in macrophage activation induced by Astragalus polysaccharide RAP. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
42
|
Jiang M, Wu YL, Li X, Zhang Y, Xia KL, Cui BW, Lian LH, Nan JX. Oligomeric proanthocyanidin derived from grape seeds inhibited NF-κB signaling in activated HSC: Involvement of JNK/ERK MAPK and PI3K/Akt pathways. Biomed Pharmacother 2017; 93:674-680. [PMID: 28692939 DOI: 10.1016/j.biopha.2017.06.105] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/14/2017] [Accepted: 06/29/2017] [Indexed: 01/05/2023] Open
|
43
|
Treatment Effects of Ischemic Stroke by Berberine, Baicalin, and Jasminoidin from Huang-Lian-Jie-Du-Decoction (HLJDD) Explored by an Integrated Metabolomics Approach. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9848594. [PMID: 28894512 PMCID: PMC5574319 DOI: 10.1155/2017/9848594] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/05/2017] [Accepted: 05/14/2017] [Indexed: 12/03/2022]
Abstract
Berberine, baicalin, and jasminoidin were major active ingredients of Huang-Lian-Jie-Du-Decoction (HLJDD), a famous prescription of traditional Chinese medicine (TCM), which has been used for the treatment of ischemic stroke. The aim of the present study was to classify their roles in the treatment effects of ischemic stroke. A rat model of middle cerebral artery occlusion (MCAO) was constructed to mimic ischemic stroke and treatment effects of berberine, baicalin, and jasminoidin, and HLJDD was assessed by neurologic deficit scoring, infarct volume, histopathology, immunohistochemistry, biochemistry, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blotting. In addition, the 1H NMR metabolomics approach was used to assess the metabolic profiles, which combined with correlation network analysis successfully revealed metabolic disorders in ischemic stroke concerning the treatment of the three principal compounds from HLJDD for the first time. The combined results suggested that berberine, baicalin, and jasminoidin are responsible for the effectiveness of HLJDD on the treatment of ischemic stroke by amelioration of abnormal metabolism and regulation of oxidative stress, neuron autophagy, and inflammatory response. This integrated metabolomics approach showed its potential in understanding the function of complex formulae and clarifying the role of its components in the overall treatment effects.
Collapse
|
44
|
Wang W, Liu J, Yang B, Ma Z, Liu G, Shen W, Zhang Y. Modulation of platelet-derived microparticles to adhesion and motility of human rheumatoid arthritis fibroblast-like synoviocytes. PLoS One 2017; 12:e0181003. [PMID: 28704431 PMCID: PMC5509257 DOI: 10.1371/journal.pone.0181003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/23/2017] [Indexed: 12/20/2022] Open
Abstract
Platelet-derived microparticles (PMPs) are closely associated with disease activity in rheumatoid arthritis (RA) and contribute to the inflammatory process. Rheumatoid arthritis fibroblast-like synoviocytes (RA-FLSs) play important roles in the progression of joint destruction. The aim of this study is to demonstrate whether PMPs affect the adhesion and motility of RA-FLSs. Our data indicated that PMPs promoted migration, invasion and adhesion to extracellular matrix (ECM) of RA-FLSs. Further study showed that PMPs up-regulated the expression of matrix metalloproteinase-1 (MMP1) and increased the level of phosphorylation of NF-κB (p-NF-κB) and Erk (p-Erk) in RA-FLSs. These results suggest that PMPs promote RA-FLSs adhesion and motility presumably by increasing MMP1 via activating Erk-mediated NF-κB pathway.
Collapse
Affiliation(s)
- Wenwen Wang
- Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
- School of Medicine, Yangzhou University, Yangzhou, P.R. China
| | - Jiahuan Liu
- Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
- School of Medicine, Yangzhou University, Yangzhou, P.R. China
| | - Binzhou Yang
- The Third People’s Hospital of Chengdu, Southwest JiaoTong University College of Medicine, Chengdu, P.R. China
| | - Zhongshuang Ma
- Department of Rheumatology, Yancheng Chengnan Hospital, Yancheng, P.R. China
| | - Guiping Liu
- Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
- School of Medicine, Yangzhou University, Yangzhou, P.R. China
| | - Weigan Shen
- Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, P.R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, P.R. China
- * E-mail: (YZ); (WS)
| | - Yu Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
- School of Medicine, Yangzhou University, Yangzhou, P.R. China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, P.R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, P.R. China
- * E-mail: (YZ); (WS)
| |
Collapse
|
45
|
A Review of the Molecular Mechanisms Underlying the Development and Progression of Cardiac Remodeling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3920195. [PMID: 28751931 PMCID: PMC5511646 DOI: 10.1155/2017/3920195] [Citation(s) in RCA: 281] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/30/2017] [Indexed: 02/07/2023]
Abstract
Pathological molecular mechanisms involved in myocardial remodeling contribute to alter the existing structure of the heart, leading to cardiac dysfunction. Among the complex signaling network that characterizes myocardial remodeling, the distinct processes are myocyte loss, cardiac hypertrophy, alteration of extracellular matrix homeostasis, fibrosis, defective autophagy, metabolic abnormalities, and mitochondrial dysfunction. Several pathophysiological stimuli, such as pressure and volume overload, trigger the remodeling cascade, a process that initially confers protection to the heart as a compensatory mechanism. Yet chronic inflammation after myocardial infarction also leads to cardiac remodeling that, when prolonged, leads to heart failure progression. Here, we review the molecular pathways involved in cardiac remodeling, with particular emphasis on those associated with myocardial infarction. A better understanding of cell signaling involved in cardiac remodeling may support the development of new therapeutic strategies towards the treatment of heart failure and reduction of cardiac complications. We will also discuss data derived from gene therapy approaches for modulating key mediators of cardiac remodeling.
Collapse
|
46
|
Shen Y, Sun J, Niu C, Yu D, Chen Z, Cong W, Geng F. Mechanistic evaluation of gastroprotective effects of Kangfuxin on ethanol-induced gastric ulcer in mice. Chem Biol Interact 2017; 273:115-124. [PMID: 28606470 DOI: 10.1016/j.cbi.2017.06.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/22/2017] [Accepted: 06/07/2017] [Indexed: 02/06/2023]
Abstract
This study was designed to evaluate the gastroprotective effect of Kangfuxin (KFX), a Chinese patent medicine constituent isolated from American cockroach, on ethanol-induced gastric ulcer in mice and to elucidate the potential mechanisms of the effect involved. According to the results, mice treated with alcohol appeared obvious gastric mucosal injury, while treatment with Cimetidine (a positive control) and KFX significantly relieved the damage, along with decreased oxidative stress and apoptosis indexes. Subsequently, we conducted a label-free quantitative proteomic (LFQ) and found that NF-κB and PI3K/AKT signaling pathway participated in gastroprotective effect of KFX. Furthermore, Western blot analysis revealed that KFX treatment inhibited the expression of TNF-α, IL-1β, greatly reduced the phosphorylation level of IκB and repressed the nuclear translocation of NF-κB p65, which demonstrated that KFX inhibited the activation of NF-κB pathway. Meanwhile, the PI3K/AKT pathway was also involved in regulating the anti-inflammation effect. These findings define for the first time that the gastroprotective effects of KFX against gastric ulcer can be attributed to its role in NF-κB inhibition.
Collapse
Affiliation(s)
- Yongmei Shen
- Sichuan Key Laboratory of Medical American Cockroach, Chengdu, China
| | - Jia Sun
- College of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Chao Niu
- College of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Dongdong Yu
- College of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Zhiwei Chen
- College of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Weitao Cong
- College of Pharmacy, Wenzhou Medical University, Wenzhou, China.
| | - Funeng Geng
- Sichuan Key Laboratory of Medical American Cockroach, Chengdu, China.
| |
Collapse
|
47
|
Progenitor Cells for Arterial Repair: Incremental Advancements towards Therapeutic Reality. Stem Cells Int 2017; 2017:8270498. [PMID: 28232850 PMCID: PMC5292398 DOI: 10.1155/2017/8270498] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 12/18/2016] [Indexed: 02/08/2023] Open
Abstract
Coronary revascularization remains the standard treatment for obstructive coronary artery disease and can be accomplished by either percutaneous coronary intervention (PCI) or coronary artery bypass graft surgery. Considerable advances have rendered PCI the most common form of revascularization and improved clinical outcomes. However, numerous challenges to modern PCI remain, namely, in-stent restenosis and stent thrombosis, underscoring the importance of understanding the vessel wall response to injury to identify targets for intervention. Among recent promising discoveries, endothelial progenitor cells (EPCs) have garnered considerable interest given an increasing appreciation of their role in vascular homeostasis and their ability to promote vascular repair after stent placement. Circulating EPC numbers have been inversely correlated with cardiovascular risk, while administration of EPCs in humans has demonstrated improved clinical outcomes. Despite these encouraging results, however, advancing EPCs as a therapeutic modality has been hampered by a fundamental roadblock: what constitutes an EPC? We review current definitions and sources of EPCs as well as the proposed mechanisms of EPC-mediated vascular repair. Additionally, we discuss the current state of EPCs as therapeutic agents, focusing on endogenous augmentation and transplantation.
Collapse
|
48
|
Chen J, Kieswich JE, Chiazza F, Moyes AJ, Gobbetti T, Purvis GSD, Salvatori DCF, Patel NSA, Perretti M, Hobbs AJ, Collino M, Yaqoob MM, Thiemermann C. IκB Kinase Inhibitor Attenuates Sepsis-Induced Cardiac Dysfunction in CKD. J Am Soc Nephrol 2017; 28:94-105. [PMID: 27153924 PMCID: PMC5198262 DOI: 10.1681/asn.2015060670] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 03/09/2016] [Indexed: 01/01/2023] Open
Abstract
Patients with CKD requiring dialysis have a higher risk of sepsis and a 100-fold higher mortality rate than the general population with sepsis. The severity of cardiac dysfunction predicts mortality in patients with sepsis. Here, we investigated the effect of preexisting CKD on cardiac function in mice with sepsis and whether inhibition of IκB kinase (IKK) reduces the cardiac dysfunction in CKD sepsis. Male C57BL/6 mice underwent 5/6 nephrectomy, and 8 weeks later, they were subjected to LPS (2 mg/kg) or sepsis by cecal ligation and puncture (CLP). Compared with sham operation, nephrectomy resulted in significant increases in urea and creatinine levels, a small (P<0.05) reduction in ejection fraction (echocardiography), and increases in the cardiac levels of phosphorylated IκBα, Akt, and extracellular signal-regulated kinase 1/2; nuclear translocation of the NF-κB subunit p65; and inducible nitric oxide synthase (iNOS) expression. When subjected to LPS or CLP, compared with sham-operated controls, CKD mice exhibited exacerbation of cardiac dysfunction and lung inflammation, greater increases in levels of plasma cytokines (TNF-α, IL-1β, IL-6, and IL-10), and greater increases in the cardiac levels of phosphorylated IKKα/β and IκBα, nuclear translocation of p65, and iNOS expression. Treatment of CKD mice with an IKK inhibitor (IKK 16; 1 mg/kg) 1 hour after CLP or LPS administration attenuated these effects. Thus, preexisting CKD aggravates the cardiac dysfunction caused by sepsis or endotoxemia in mice; this effect may be caused by increased cardiac NF-κB activation and iNOS expression.
Collapse
Affiliation(s)
- Jianmin Chen
- Center for Translational Medicine and Therapeutics and
| | | | - Fausto Chiazza
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Amie J Moyes
- Center for Translational Medicine and Therapeutics and
| | - Thomas Gobbetti
- Center for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Daniela C F Salvatori
- Central Laboratory Animal Facility, Leiden University Medical Center, Leiden, The Netherlands; and
| | | | - Mauro Perretti
- Center for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Muhammad M Yaqoob
- Center for Translational Medicine and Therapeutics and
- Department of Renal Medicine and Transplantation, Royal London Hospital, Whitechapel, London, United Kingdom
| | | |
Collapse
|
49
|
Pellino-1 promotes lung carcinogenesis via the stabilization of Slug and Snail through K63-mediated polyubiquitination. Cell Death Differ 2016; 24:469-480. [PMID: 28009353 PMCID: PMC5457685 DOI: 10.1038/cdd.2016.143] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 10/26/2016] [Accepted: 11/08/2016] [Indexed: 12/20/2022] Open
Abstract
Pellino-1 is an E3 ubiquitin ligase acting as a critical mediator for a variety of immune receptor signaling pathways, including Toll-like receptors, interleukin-1 receptor and T-cell receptors. We recently showed that the Pellino-1-transgenic (Tg) mice developed multiple tumors with different subtypes in hematolymphoid and solid organs. However, the molecular mechanism underlying the oncogenic role of Pellino-1 in solid tumors remains unknown. Pellino-1-Tg mice developed adenocarcinoma in the lungs, and Pellino-1 expression was higher in human lung adenocarcinoma cell lines compared with non-neoplastic bronchial epithelial cell lines. Pellino-1 overexpression increased the cell proliferation, survival, colony formation, invasion and migration of lung adenocarcinoma cells, whereas Pellino-1 knock-down showed the opposite effect. Pellino-1 overexpression activated PI3K/Akt and ERK signaling pathways and elicited an epithelial–mesenchymal transition (EMT) phenotype of lung adenocarcinoma cells. Pellino-1-mediated EMT was demonstrated through morphology, the upregulation of Vimentin, Slug and Snail expression and the downregulation of E-cadherin and β-catenin expression. Notably, Pellino-1 had a direct effect on the overexpression of Snail and Slug through Lys63-mediated polyubiquitination and the subsequent stabilization of these proteins. Pellino-1 expression level was significantly correlated with Snail and Slug expression in human lung adenocarcinoma tissues, and lung tumors from Pellino-1-Tg mice showed Snail and Slug overexpression. The Pellino-1-mediated increase in the migration of lung adenocarcinoma cells was mediated by Snail and Slug expression. Taken together, these results show that Pellino-1 contributes to lung tumorigenesis by inducing overexpression of Snail and Slug and promoting EMT. Pellino-1 might be a potential therapeutic target for lung cancer.
Collapse
|
50
|
BACE1-Deficient Mice Exhibit Alterations in Immune System Pathways. Mol Neurobiol 2016; 55:709-717. [PMID: 28004339 DOI: 10.1007/s12035-016-0341-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 12/02/2016] [Indexed: 01/09/2023]
Abstract
BACE1 encodes for the beta-site amyloid precursor protein cleaving enzyme 1 or β-secretase. Genetic deletion of Bace1 leads to behavioral alterations and affects midbrain dopaminergic signaling and memory processes. In order to further understand the role of BACE1 in brain function and behavior, we performed microarray transcriptome profiling and gene pathway analysis in the hippocampus of BACE1-deficient mice compared to wild type. We identified a total of 91 differentially expressed genes (DEGs), mostly enriched in pathways related to the immune and inflammation systems, particularly IL-9 and NF-κB activation pathways. Serum levels of IL-9 were elevated in BACE1-deficient mice. Our network analysis supports an intimate connection between immune response via NF-κB and BACE1 signaling through the NRG1/Akt1 pathway. Our findings warrant future mechanistic studies to determine if BACE1 signaling and the IL-9 pathway interact to alter behavior and brain function. This study opens new avenues in the investigation of hippocampus-related neuroimmunological and neuroinflammation-associated disorders.
Collapse
|