1
|
Guo Z, Lv X, Li J, Yue S, Du J. Blastocyst Cavity Expansion Promotes Cell Polarization During Early Development of Mouse Embryos. Birth Defects Res 2025; 117:e2484. [PMID: 40347064 DOI: 10.1002/bdr2.2484] [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: 10/11/2024] [Revised: 04/27/2025] [Accepted: 04/30/2025] [Indexed: 05/12/2025]
Abstract
BACKGROUND Cell polarization is an important morphological process that is crucial for the formation and function of tissues and organs. The blastocyst cavity expansion is an apparent event during the second cell fate specification in mouse embryos, yet its impact on cell polarization remains unclear. In this study, we investigate the effects of blastocyst cavity expansion on cell polarization. METHODS The methods of this study involve hyperosmotic treatment or disruption of TE cortical tension by laser ablation, combined with immunofluorescence. RESULTS We found that inhibition of the blastocyst cavity expansion through hypertonic treatment or disruption of TE cortical tension by laser ablation suppresses the levels of the ζ isotype of protein kinase C (PKC ζ) which is a member of the atypical PKC subfamily involved in cell polarization. We further found that during the embryonic stages E3.5 to E4.0, the expression of extracellular signal-regulated kinase 1 (ERK1), a key upstream regulator of PKC ζ, is altered in a similar tendency to that of PKC ζ, indicating a potential regulatory function of ERK1 in cell polarization during early development of mouse embryos. CONCLUSIONS This study reveals the function of the mechanical behavior of embryos in cell polarization of early mammalian embryos. The relationship between cell polarization and blastocyst cavity expansion in early embryonic development provides a new understanding, thereby offering fresh insights for the screening and detection of indicators for normal blastocyst development.
Collapse
Affiliation(s)
- Zheng Guo
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xinxin Lv
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jianwen Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shiping Yue
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jing Du
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
2
|
Weisheng Z, Nakayama J, Inomata Y, Higashiyama S, Hiratsuka T. A sensitive ERK fluorescent probe reveals the significance of minimal EGF-induced transcription. Cell Struct Funct 2025; 50:15-24. [PMID: 39694501 DOI: 10.1247/csf.24070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
Abstract
Extracellular signal-regulated kinase (ERK) regulates multiple cellular functions through distinct activation patterns. Genetically encoded fluorescent probes are instrumental in dissecting the ERK activity dynamics in living cells. Here we modified a previously reported Förster resonance energy transfer (FRET) probe for ERK, EKAREN5 by replacing its mTurquoise2 and YPet sequences with mTurquoise-GL and a synonymous codon variant of YPet, respectively. The modified biosensor, EKAREN5-gl, showed an increased sensitivity to EGF-induced ERK activation responding to a very low dose (20 pg/ml) of EGF stimulation. We quantitatively characterized two FRET-based ERK probes, EKAREN5 and EKAREN5-gl, and a subcellular kinase translocation-based probe, ERK-KTR. We found the three biosensors differently respond to EGF stimulations with different intensity, duration, and latency. Furthermore, we investigated how the minimal EGF-induced ERK activation affects the downstream transcription in HeLa cells by comprehensive transcriptional analysis. We found the minimal ERK activation leads to a distinct transcriptional pattern from those induced by higher ERK activations. Our study highlights the significance of sensitive fluorescent probes to understand cellular signal dynamics and the role of minimal ERK activation in regulating transcription.Key words: fluorescent probe, ERK, FRET, KTR.
Collapse
Affiliation(s)
- Zhang Weisheng
- Department of Molecular Oncology, Graduate School of Medicine, Osaka University
- Department of Oncogenesis and Growth Regulation, Research Center, Osaka International Cancer Institute
| | - Jun Nakayama
- Department of Oncogenesis and Growth Regulation, Research Center, Osaka International Cancer Institute
| | - Yukino Inomata
- Department of Oncogenesis and Growth Regulation, Research Center, Osaka International Cancer Institute
| | - Shigeki Higashiyama
- Department of Oncogenesis and Growth Regulation, Research Center, Osaka International Cancer Institute
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS)
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine
| | - Toru Hiratsuka
- Department of Molecular Oncology, Graduate School of Medicine, Osaka University
- Department of Oncogenesis and Growth Regulation, Research Center, Osaka International Cancer Institute
| |
Collapse
|
3
|
Yuan CY, Zuo L, Dong YC, Liu BX, Qi H. Secretogranin III: a promising therapeutic target for intraocular neovascular lesions. Int Ophthalmol 2025; 45:26. [PMID: 39832055 PMCID: PMC11746947 DOI: 10.1007/s10792-024-03393-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE The purpose of this study is to investigate the role of Secretogranin III (Scg3) in the pathogenesis of intraocular neovascular diseases and assess its potential as a therapeutic target for novel treatment strategies. METHODS A literature review was conducted to examine the expression of Scg3 in intraocular neovascular diseases. We reviewed studies on the interaction of Scg3 with its homologous receptors and its effect on endothelial cell proliferation, migration, and vascular permeability-key processes involved in angiogenesis and neovascularization. RESULTS Scg3 was found to be upregulated in the tissues affected by diabetic retinopathy (DR), retinopathy of prematurity (ROP), and choroidal neovascularization. In DR, Scg3 expression was linked to retinal neovascularization, where it facilitated endothelial cell proliferation and migration, essential processes for the formation of new blood vessels. Similarly, in ROP, Scg3 was associated with fibrovascular tissue proliferation within avascular retinal zones, contributing to the pathological neovascularization seen in premature infants. In the context of age-related macular degeneration (AMD), Scg3 appeared to play a role in choroidal neovascularization, where it promoted the invasion of choroidal capillaries into the retinal pigment epithelium. Furthermore, Scg3's binding to its homologous receptors was shown to enhance vascular permeability, potentially exacerbating fluid leakage and edema in these diseases, which is a hallmark of exudative conditions. Collectively, these findings suggest that Scg3 plays a pivotal role in driving angiogenesis and vascular permeability in intraocular neovascular diseases CONCLUSION: The upregulation of Scg3 in DR, ROP, and choroidal neovascularization highlights its potential as a novel therapeutic target. Inhibition of Scg3 could offer a new avenue for treating these sight-threatening conditions.
Collapse
Affiliation(s)
- Chao-Yi Yuan
- Department of Ophthalmology, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, 130041, Jilin, China
| | - Ling Zuo
- Department of Ophthalmology, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, 130041, Jilin, China
| | - Yu-Chen Dong
- Department of Ophthalmology, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, 130041, Jilin, China
| | - Bao-Xing Liu
- Department of Ophthalmology, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, 130041, Jilin, China
| | - Hui Qi
- Department of Ophthalmology, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, 130041, Jilin, China.
| |
Collapse
|
4
|
Zhao Y, Zhang J, Zhang G, Huang H, Tan WS, Cai H. Injectable Nanocomposite Hydrogel with Synergistic Biofilm Eradication and Enhanced Re-epithelialization for Accelerated Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:69086-69102. [PMID: 39635909 DOI: 10.1021/acsami.4c17855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Diabetic wounds remain a critical clinical challenge due to their harsh microenvironment, which impairs cellular function, hinders re-epithelialization and tissue remodeling, and slows healing. Injectable nanocomposite hydrogel dressings offer a promising strategy for diabetic wound repair. In this study, we developed an injectable nanocomposite hydrogel dressing (HDL@W379) using LAP@W379 nanoparticles and an injectable hyaluronic acid-based hydrogel (HA-ADH-ODEX). This dressing provided a sustained, pH-responsive release of W379 antimicrobial peptides, effectively regulating the wound microenvironment to enhance healing. The HDL@W379 hydrogel featured multifunctional properties, including mechanical stability, injectability, self-healing, biocompatibility, and tissue adhesion. In vitro, the HDL@W379 hydrogel achieved synergistic biofilm elimination and subsequent activation of basal cell migration and endothelial cell tube formation. Pathway analysis indicated that the HDL@W379 hydrogel enhances basal cell migration through MEK/ERK pathway activation. In methicillin-resistant Staphylococcus aureus (MRSA)-infected diabetic wounds, the HDL@W379 hydrogel accelerated wound healing by inhibiting bacterial proliferation and promoting re-epithelialization, regenerating the granulation tissue, enhancing collagen deposition, and facilitating angiogenesis. Overall, this strategy of biofilm elimination and basal cell activation to continuously regulate the diabetic wound microenvironment offers an innovative approach to treating chronic wounds.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Jingwei Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Guofeng Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Huimin Huang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| |
Collapse
|
5
|
Feng Y, Tu SQ, Hou YL, Shao YT, Chen L, Mai ZH, Wang YX, Wei JM, Zhang S, Ai H, Chen Z. Alendronate sodium induces G1 phase arrest and apoptosis in human umbilical vein endothelial cells by inhibiting ROS-mediated ERK1/2 signaling. Toxicology 2024; 508:153917. [PMID: 39137827 DOI: 10.1016/j.tox.2024.153917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
Bisphosphonates are potent bone resorption inhibitors, among which alendronate sodium (ALN) is commonly prescribed for most osteoporosis patients, but long-term application of ALN can cause bisphosphonate-related osteonecrosis of jaw (BRONJ), the pathogenesis of which remains unclear. Previous studies have suggested that bisphosphonates cause jaw ischemia by affecting the biological behavior of vascular endothelial cells, leading to BRONJ. However, the impacts of ALN on vascular endothelial cells and its mechanism remain unclear. The purpose of this work is to assess the influence of ALN on human umbilical vein endothelial cells (HUVECs) and clarify the molecular pathways involved. We found that high concentration of ALN induced G1 phase arrest in HUVECs, demonstrated by downregulation of Cyclin D1 and Cyclin D3. Moreover, high concentration of ALN treatment showed pro-apoptotic effect on HUVECs, demonstrated by increased levels of the cleaved caspase-3, the cleaved PARP and Bax, along with decreased levels of anti-apoptotic protein Bcl-2. Further experiments showed that ERK1/2 phosphorylation was decreased. Additionally, ALN provoked the build-up of reactive oxygen species (ROS) in HUVECs, leading to ERK1/2 pathway suppression. N-acetyl-L-cysteine (NAC), a ROS scavenger, efficiently promoted the ERK1/2 phosphorylation and mitigated the G1 phase arrest and apoptosis triggered by ALN in HUVECs. PD0325901, an inhibitor of ERK1/2 that diminishes the ERK1/2 phosphorylation enhanced the ALN-induced G1 phase arrest and apoptosis in HUVECs. These findings show that ALN induces G1 phase arrest and apoptosis through ROS-mediated ERK1/2 pathway inhibition in HUVECs, providing novel insights into the pathogenic process, prevention and treatment of BRONJ in individuals receiving extended use of ALN.
Collapse
Affiliation(s)
- Yi Feng
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shao-Qin Tu
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu-Luan Hou
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yi-Ting Shao
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lin Chen
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhi-Hui Mai
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu-Xuan Wang
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Department of Stomatology, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, China
| | - Jia-Ming Wei
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sai Zhang
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hong Ai
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Zheng Chen
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
6
|
Kundu D, Acharya S, Wang S, Kim KM. Unveiling the intracellular dynamics of α4β2 nAChR-mediated ERK activation through the interplay of arrestin, Gβγ, and PKCβII. Life Sci 2024; 355:122994. [PMID: 39163903 DOI: 10.1016/j.lfs.2024.122994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/28/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024]
Abstract
AIMS In contrast to G protein-coupled receptors or receptor tyrosine kinases, the mechanism underlying ERK activation through nicotine acetylcholine receptors (nAChRs), members of the ligand-gated ion channel family, remains poorly elucidated. This study aimed to delineate the signaling pathway responsible for ERK activation by the α4β2 nAChR subtype, which is implicated in nicotine addiction and various mental disorders. MATERIALS AND METHODS Loss-of-function strategies and mutants of arrestin2/PKCβII with distinct functional characteristics were employed to identify the cellular components and processes involved in ERK activation. KEY FINDINGS ERK activation via α4β2 nAChR was observed within the nucleus and necessitated the nuclear translocation of arrestin2 and PKCβII, which exhibited mutual augmentation. Activation of PKCβII by α4β2 nAChR stimulation facilitated the nuclear translocation of arrestin2 by enhancing its interaction with importin β1. Apart from scaffolding ERK activation in the nucleus, arrestin2, in cooperation with GRK2, facilitated the activation of the Src/Syk/PKCβII signaling cascade, leading to the nuclear entry of PKCβII in a Gβγ-dependent manner. Upon nuclear localization, PKCβII underwent ubiquitination by Mdm2 and interacted with MEK1, resulting in ERK activation. In summary, α4β2 nAChR-mediated ERK activation in the nucleus involves the nuclear translocation of arrestin2 and PKCβII, which is reciprocally facilitated via positive feedback augmentation. SIGNIFICANCE As α4β2 nAChRs play a pivotal role in various cellular processes including drug addiction and mental disorders, our findings will offer insights into understanding the pathogenesis of α4β2 nAChR-related disorders and may facilitate the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Dooti Kundu
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Srijan Acharya
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Shujie Wang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
7
|
Xin J, Song M, Liu X, Zou H, Wang J, Xiao L, Jia Y, Zhang G, Jiang W, Lei M, Yang Y, Jiang Y. A new strategy of using low-dose caffeic acid carbon nanodots for high resistance to poorly differentiated human papillary thyroid cancer. J Nanobiotechnology 2024; 22:571. [PMID: 39294724 PMCID: PMC11409714 DOI: 10.1186/s12951-024-02792-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024] Open
Abstract
Thyroid cancer is one of the most common endocrine malignancies in clinical practice. Traditional surgery and radioactive iodine ablation have poor treatment results for poorly differentiated thyroid cancer, and there is a risk of metastasis and recurrence. In this study, caffeic acid, a natural herbal extract with certain biological activity, has been as precursor to prepare new caffeic acid carbon nanodots via a one-step hydrothermal method. The caffeic acid carbon nanodots retains part of the structure and biological activity of caffeic acid, and have good biocompatibility, water solubility and stability. The construction of the carbon nanodots could effectively improve their bio-absorption rate and the efficacy. In vitro cell experiments showed that low-dose caffeic acid carbon nanodots had a significant inhibitory effect on poorly differentiated papillary thyroid carcinoma BCPAP cells. At low concentrations of 16 µg/mL, the inhibition rate of human thyroid cancer cells BCPAP was ~ 79%. The anti-tumor mechanism was predicted and verified by transcriptome, real-time quantitative PCR and western blot experiments. The caffeic acid carbon nanodots showed to simultaneously downregulate the expression of KRAS, p-BRAF, p-MEK1 and p-ERK1/2, the four continuous key proteins in a MAPK classical signaling pathway. In vivo experiments further confirmed the caffeic acid carbon nanodots could significantly inhibit the tumorigenicity of xenografts in papillary thyroid carcinoma at quite low doses. This piece of work provides a new nanomedicine and therapeutic strategy for highly resistant poorly differentiated papillary thyroid carcinoma.
Collapse
Affiliation(s)
- Jingwei Xin
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China
- Jilin Provincial Key Laboratory of Surgical Translational Medicine, Division of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Meiwei Song
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Xiangling Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Hongrui Zou
- Jilin Provincial Key Laboratory of Surgical Translational Medicine, Division of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Jifeng Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Lizhi Xiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yunxiao Jia
- Department Gynecol & Obstet, Changchun Obstet Gynecol Hospital, Changchun Women and Children Health Hospital, Changchun, 130042, China.
| | - Guoqi Zhang
- Harvard Medical School, Bonston Children's Hospital, Bonston, 02111, US
| | - Wei Jiang
- Department Gynecol & Obstet, Changchun Obstet Gynecol Hospital, Changchun Women and Children Health Hospital, Changchun, 130042, China
| | - Ming Lei
- Department Gynecol & Obstet, Changchun Obstet Gynecol Hospital, Changchun Women and Children Health Hospital, Changchun, 130042, China
| | - Yanyan Yang
- Jilin Provincial Key Laboratory of Surgical Translational Medicine, Division of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Yingnan Jiang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
- Australian Institute for Bioengineering and Nanotechnology , The University of Queensland, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
8
|
Hossain MA. Targeting the RAS upstream and downstream signaling pathway for cancer treatment. Eur J Pharmacol 2024; 979:176727. [PMID: 38866361 DOI: 10.1016/j.ejphar.2024.176727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Cancer often involves the overactivation of RAS/RAF/MEK/ERK (MAPK) and PI3K-Akt-mTOR pathways due to mutations in genes like RAS, RAF, PTEN, and PIK3CA. Various strategies are employed to address the overactivation of these pathways, among which targeted therapy emerges as a promising approach. Directly targeting specific proteins, leads to encouraging results in cancer treatment. For instance, RTK inhibitors such as imatinib and afatinib selectively target these receptors, hindering ligand binding and reducing signaling initiation. These inhibitors have shown potent efficacy against Non-Small Cell Lung Cancer. Other inhibitors, like lonafarnib targeting Farnesyltransferase and GGTI 2418 targeting geranylgeranyl Transferase, disrupt post-translational modifications of proteins. Additionally, inhibition of proteins like SOS, SH2 domain, and Ras demonstrate promising anti-tumor activity both in vivo and in vitro. Targeting downstream components with RAF inhibitors such as vemurafenib, dabrafenib, and sorafenib, along with MEK inhibitors like trametinib and binimetinib, has shown promising outcomes in treating cancers with BRAF-V600E mutations, including myeloma, colorectal, and thyroid cancers. Furthermore, inhibitors of PI3K (e.g., apitolisib, copanlisib), AKT (e.g., ipatasertib, perifosine), and mTOR (e.g., sirolimus, temsirolimus) exhibit promising efficacy against various cancers such as Invasive Breast Cancer, Lymphoma, Neoplasms, and Hematological malignancies. This review offers an overview of small molecule inhibitors targeting specific proteins within the RAS upstream and downstream signaling pathways in cancer.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
9
|
Huang W, Zhou H, He Y, Wang A, Wang B, Chen Y, Liu C, Wang H, Xie W, Kong H. A novel PDGFR inhibitor WQ-C-401 prevents pulmonary vascular remodeling in rats with monocrotaline-induced pulmonary arterial hypertension. Exp Cell Res 2024; 441:114154. [PMID: 38996959 DOI: 10.1016/j.yexcr.2024.114154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Platelet-derived growth factor (PDGF) is one of the most important cytokines associated with pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). PDGF receptor (PDGFR) inhibition exerted therapeutic effects on PAH in clinical trials, but serious side effects warrant the withdrawal of existing drugs. In this study, a novel highly selective PDGFR inhibitor WQ-C-401 was developed, and its effects on PDGFR signaling pathway and pulmonary vascular remodeling in PAH were investigated. Cell proliferation assays and Western blot analysis of PDGFRα/β phosphorylation showed that WQ-C-401 inhibited PDGFR-mediated cell proliferation assay and suppressed PDGFR phosphorylation in a concentration-dependent manner. DiscoverX's KinomeScanTM technology confirmed the good kinome selectivity of WQ-C-401 (S score (1) of PDGFR = (0.01)). In monocrotaline (MCT)-induced PAH rats, intragastric administration of WQ-C-401 (25, 50, 100 mg/kg/d) or imatinib (50 mg/kg/d, positive control) significantly decreased right ventricular systolic pressure (RVSP). Histological analysis demonstrated that WQ-C-401 inhibited pulmonary vascular remodeling by reducing muscularization and fibrosis, as well as alleviated right ventricular hypertrophy in MCT-treated rats. In addition, WQ-C-401 suppressed MCT-induced cell hyperproliferation and CD68+ macrophage infiltration around the pulmonary artery. In vitro, WQ-C-401 inhibited PDGF-BB-induced proliferation and migration of human pulmonary arterial smooth muscle cells (PASMCs). Moreover, Western blot analysis showed that WQ-C-401 concertration-dependently inhibited PDGF-BB-induced phosphorylation of ERK1/2 and PDGFRβ Y751, decreased collagen Ⅰ synthesis and increased alpha smooth muscle actin (α-SMA) expression in PASMCs. Collectively, our results suggest that WQ-C-401 is a selective and potent PDGFR inhibitor which could be a promising drug for the therapeutics of PAH by preventing pulmonary vascular remodeling.
Collapse
MESH Headings
- Animals
- Monocrotaline
- Vascular Remodeling/drug effects
- Rats
- Cell Proliferation/drug effects
- Male
- Rats, Sprague-Dawley
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/chemically induced
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/pathology
- Humans
- Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors
- Receptors, Platelet-Derived Growth Factor/metabolism
- Phosphorylation/drug effects
- Pulmonary Artery/drug effects
- Pulmonary Artery/pathology
- Pulmonary Artery/metabolism
- Signal Transduction/drug effects
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/prevention & control
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/metabolism
- Protein Kinase Inhibitors/pharmacology
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
Collapse
Affiliation(s)
- Wen Huang
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Hong Zhou
- Department of Pulmonary & Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, Jiangsu, PR China
| | - Yiting He
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Aoli Wang
- Anhui Province Key Laboratory of Medical Physics & Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China
| | - Beilei Wang
- Anhui Province Key Laboratory of Medical Physics & Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China
| | - Yongfei Chen
- Anhui Province Key Laboratory of Medical Physics & Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China
| | - Chenyang Liu
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Hong Wang
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Weiping Xie
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China.
| | - Hui Kong
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China.
| |
Collapse
|
10
|
Márton A, Veres KB, Erdődi F, Udvardy M, Illés Á, Rejtő L. The roles of phosphorylation of signaling proteins in the prognosis of acute myeloid leukemia. Pathol Oncol Res 2024; 30:1611747. [PMID: 39035053 PMCID: PMC11257863 DOI: 10.3389/pore.2024.1611747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/12/2024] [Indexed: 07/23/2024]
Abstract
Signaling pathways of Retinoblastoma (Rb) protein, Akt-kinase, and Erk-kinase (extracellular signal-regulated kinase) have an important role in the pathogenesis of acute myeloid leukemia. Constitutive activation of these proteins by phosphorylation contributes to cell survival by regulation of cell cycle, proliferation and proapoptotic signaling processes. According to previous data phosphorylated forms of these proteins represent a worse outcome for cancer patients. We investigated the presence of phosphorylated Rb (P-Rb), Akt (P-Akt) and Erk (P-Erk) proteins by Western blot technique using phospho-specific antibodies in bone marrow or peripheral blood samples of 69 AML patients, 36 patients with myelodysplastic syndrome (MDS) and 10 healthy volunteers. Expression level of PTEN (Phosphatase and tensin homolog) and PHLPP (PH domain and leucine-rich repeat Protein Phosphatase) phosphatases, the negative regulators of Akt kinase pathway were also examined. We tested the effect of these proteins on survival and on the correlation with known prognostic features in AML. We found 46.3% of AML patients had detectable P-Rb, 34.7% had P-Akt and 28.9% had P-Erk protein. 66.1% of patients expressing PTEN, 38.9% PHLPP, 37.2% both PTEN and PHLPP and 32.2% neither PTEN nor PHLPP phosphatases. Compared to nucleophosmin mutation (NPMc) negative samples P-Erk was significantly less in nucleophosmin mutated patients, P-Rb was significantly less in patients' group with more than 30 G/L peripheral leukocyte count by diagnosis. PHLPP was significantly present in FAB type M5. The expression of P-Rb represented significant better overall survival (OS), while P-Akt represented significantly worse event-free survival (EFS) in unfavorable cytogenetics patients. The presence of both PHLPP and PTEN phosphatases contributes to better OS and EFS, although the differences were not statistically significant. We confirmed significant positive correlation between P-Akt and PHLPP. Assessing the phosphorylation of Rb, Akt and Erk may define a subgroup of AML patients who would benefit especially from new targeted treatment options complemented the standard chemotherapy, and it may contribute to monitoring remission, relapse or progression of AML.
Collapse
Affiliation(s)
- Adrienn Márton
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | | | - Ferenc Erdődi
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklós Udvardy
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Árpád Illés
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Rejtő
- Department of Hematology, Szabolcs-Szatmár-Bereg County Teaching Hospital, Nyíregyháza, Hungary
| |
Collapse
|
11
|
Rao J, Wang T, Wang K, Qiu F. Integrative analysis of metabolomics and proteomics reveals mechanism of berberrubine-induced nephrotoxicity. Toxicol Appl Pharmacol 2024; 488:116992. [PMID: 38843998 DOI: 10.1016/j.taap.2024.116992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Berberrubine (BRB), a main metabolite of berberine, has stronger hypoglycemic and lipid-lowering activity than its parent form. We previously found that BRB could cause obvious nephrotoxicity, but the molecular mechanism involved remains unknown. In this study, we systematically integrated metabolomics and quantitative proteomics to reveal the potential mechanism of nephrotoxicity caused by BRB. Metabolomic analysis revealed that 103 significant- differentially metabolites were changed. Among the mentioned compounds, significantly upregulated metabolites were observed for phosphorylcholine, sn-glycerol-3-phosphoethanolamine, and phosphatidylcholine. The top three enriched KEGG pathways were the mTOR signaling pathway, central carbon metabolism in cancer, and choline metabolism in cancer. ERK1/2 plays key roles in all three metabolic pathways. To further confirm the main signaling pathways involved, a proteomic analysis was conducted to screen for key proteins (such as Mapk1, Mapk14, and Caspase), indicating the potential involvement of cellular growth and apoptosis. Moreover, combined metabolomics and proteomics analyses revealed the participation of ERK1/2 in multiple metabolic pathways. These findings indicated that ERK1/2 regulated the significant- differentially abundant metabolites determined via metabolomics analysis. Notably, through a cellular thermal shift assay (CETSA) and molecular docking, ERK1/2 were revealed to be the direct binding target involved in BRB-induced nephrotoxicity. To summarize, this study sheds light on the understanding of severe nephrotoxicity caused by BRB and provides scientific basis for its safe use and rational development.
Collapse
Affiliation(s)
- Jinqiu Rao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Tianwang Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Kai Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
12
|
Ge B, Yan K, Sang R, Wang W, Liu X, Yu M, Liu X, Qiu Q, Zhang X. Integrated network toxicology, molecular docking, and in vivo experiments to elucidate molecular mechanism of aflatoxin B1 hepatotoxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 275:116278. [PMID: 38564860 DOI: 10.1016/j.ecoenv.2024.116278] [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: 01/11/2024] [Revised: 03/07/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Due to the rise in temperature and sea level caused by climate change, the detection rate of aflatoxin B1 (AFB1) in food crops has increased dramatically, and the frequency and severity of aflatoxicosis in humans and animals are also increasing. AFB1 has strong hepatotoxicity, causing severe liver damage and even cancer. However, the mechanism of AFB1 hepatotoxicity remains unclear. By integrating network toxicology, molecular docking and in vivo experiments, this research was designed to explore the potential hepatotoxicity mechanisms of AFB1. Thirty-three intersection targets for AFB1-induced liver damage were identified using online databases. PI3K/AKT1, MAPK, FOXO1 signaling pathways, and apoptosis were significantly enriched. In addition, the proteins of ALB, AKT1, PIK3CG, MAPK8, HSP90AA1, PPARA, MAPK1, EGFR, FOXO1, and IGF1 exhibited good affinity with AFB1. In vivo experiments, significant pathological changes occurred in the liver of mice. AFB1 induction increased the expression levels of EGFR, ERK, and FOXO1, and decreased the expression levsls of PI3K and AKT1. Moreover, AFB1 treatment caused an increase in Caspase3 expression, and a decrease in Bcl2/Bax ratio. By combining network toxicology with in vivo experiments, this study confirms for the first time that AFB1 promotes the FOXO1 signaling pathway by inactivating PI3K/AKT1 and activating EGFR/ERK signaling pathways, hence aggravating hepatocyte apoptosis. This research provides new strategies for studying the toxicity of environmental pollutants and new possible targets for the development of hepatoprotective drugs.
Collapse
Affiliation(s)
- Bingjie Ge
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Kexin Yan
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Rui Sang
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Wei Wang
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Xinman Liu
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Minghong Yu
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Xiaotong Liu
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Qian Qiu
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Xuemei Zhang
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China.
| |
Collapse
|
13
|
Oria RS, Anyanwu GE, Nto JN, Ikpa JO. Curcumin abrogates cobalt-induced neuroinflammation by suppressing proinflammatory cytokines release, inhibiting microgliosis and modulation of ERK/MAPK signaling pathway. J Chem Neuroanat 2024; 137:102402. [PMID: 38428651 DOI: 10.1016/j.jchemneu.2024.102402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024]
Abstract
Curcumin, a bioactive polyphenol derived from turmeric, has been reported to have anti-inflammatory properties. The current study investigated the anti-inflammatory effect of curcumin in the hippocampal subfields (CA1 and CA3) after exposure to cobalt (Co) and the impact of ERK protein. Twenty-eight albino Wistar rats were divided into four groups, each with seven randomly selected rats as follows: Control (distilled water), Cobalt (Co) only (40 mg/kg), 120 mg/kg or 240 mg/kg curcumin + Co (40 mg/kg). Treatment was via oral gavage for 28 days. We performed a biochemical investigation to determine the levels of proinflammatory cytokines (TNFα and IL-1β). Furthermore, we conducted an immunohistochemical evaluation to assess the expression of IBA1 by microglial cells and the immunoexpression of ERK protein in the hippocampus. Results revealed a significant (p<0.05) elevation in the tissue level of TNFα and IL-1β, an increase in the number of IBA1-positive microglia, and upregulation of ERK protein in the hippocampal subfields of the rats after exposure to cobalt-only. Nevertheless, pretreatment with curcumin restored these parameters to levels comparable to control. In conclusion, our results showed that curcumin abrogated the Co-induced neuroinflammation by suppressing the release of proinflammatory biomarkers, reducing microgliosis, and modulating the ERK/MAPK pathway.
Collapse
Affiliation(s)
- Rademene S Oria
- Department of Anatomy, Faculty Of Basic Medical Sciences, University of Cross River State (UNICROSS), Cross River State, Nigeria; Department Of Anatomy, Faculty Of Basic Medical Sciences, College Of Medicine, University Of Nigeria Enugu Campus,, Enugu, Nigeria.
| | - Godson E Anyanwu
- Department Of Anatomy, Faculty Of Basic Medical Sciences, College Of Medicine, University Of Nigeria Enugu Campus,, Enugu, Nigeria; Department of Anatomy, Faculty of Biomedical Sciences, Kampala International University, Uganda
| | - Johnson N Nto
- Department Of Anatomy, Faculty Of Basic Medical Sciences, College Of Medicine, University Of Nigeria Enugu Campus,, Enugu, Nigeria
| | - James O Ikpa
- Department of Anatomy, Faculty Of Basic Medical Sciences, University of Cross River State (UNICROSS), Cross River State, Nigeria
| |
Collapse
|
14
|
Bahar ME, Kim HJ, Kim DR. Targeting the RAS/RAF/MAPK pathway for cancer therapy: from mechanism to clinical studies. Signal Transduct Target Ther 2023; 8:455. [PMID: 38105263 PMCID: PMC10725898 DOI: 10.1038/s41392-023-01705-z] [Citation(s) in RCA: 203] [Impact Index Per Article: 101.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/03/2023] [Accepted: 11/12/2023] [Indexed: 12/19/2023] Open
Abstract
Metastatic dissemination of solid tumors, a leading cause of cancer-related mortality, underscores the urgent need for enhanced insights into the molecular and cellular mechanisms underlying metastasis, chemoresistance, and the mechanistic backgrounds of individuals whose cancers are prone to migration. The most prevalent signaling cascade governed by multi-kinase inhibitors is the mitogen-activated protein kinase (MAPK) pathway, encompassing the RAS-RAF-MAPK kinase (MEK)-extracellular signal-related kinase (ERK) pathway. RAF kinase is a primary mediator of the MAPK pathway, responsible for the sequential activation of downstream targets, such as MEK and the transcription factor ERK, which control numerous cellular and physiological processes, including organism development, cell cycle control, cell proliferation and differentiation, cell survival, and death. Defects in this signaling cascade are associated with diseases such as cancer. RAF inhibitors (RAFi) combined with MEK blockers represent an FDA-approved therapeutic strategy for numerous RAF-mutant cancers, including melanoma, non-small cell lung carcinoma, and thyroid cancer. However, the development of therapy resistance by cancer cells remains an important barrier. Autophagy, an intracellular lysosome-dependent catabolic recycling process, plays a critical role in the development of RAFi resistance in cancer. Thus, targeting RAF and autophagy could be novel treatment strategies for RAF-mutant cancers. In this review, we delve deeper into the mechanistic insights surrounding RAF kinase signaling in tumorigenesis and RAFi-resistance. Furthermore, we explore and discuss the ongoing development of next-generation RAF inhibitors with enhanced therapeutic profiles. Additionally, this review sheds light on the functional interplay between RAF-targeted therapies and autophagy in cancer.
Collapse
Affiliation(s)
- Md Entaz Bahar
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea.
| |
Collapse
|
15
|
Dubourg V, Schwerdt G, Schreier B, Kopf M, Mildenberger S, Benndorf RA, Gekle M. Transcriptional impact of EGFR activation in human female vascular smooth muscle cells. iScience 2023; 26:108286. [PMID: 38026216 PMCID: PMC10651680 DOI: 10.1016/j.isci.2023.108286] [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: 06/14/2023] [Revised: 09/19/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Vascular smooth muscle cells (VSMC) are critical for the vascular tone, but they can also drive the development of vascular diseases when they lose their contractile phenotype and de-differentiate. Previous studies showed that the epidermal growth factor receptor (EGFR) of VSMC is critical for vascular health, but most of the underlying mechanisms by which VSMC-EGFR controls vascular fate have remained unknown. We combined RNA-sequencing and bioinformatics analysis to characterize the effect of EGFR-activation on the transcriptome of human primary VSMC (from different female donors) and to identify potentially affected cellular processes. Our results indicate that the activation of human VSMC-EGFR is sufficient to trigger a phenotypical switch toward a proliferative and inflammatory phenotype. The extent of this effect is nonetheless partly donor-dependent. Our hypothesis-generating study thus provides a first insight into mechanisms that could partly explain variable susceptibilities to vascular diseases in between individuals.
Collapse
Affiliation(s)
- Virginie Dubourg
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Gerald Schwerdt
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Barbara Schreier
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Michael Kopf
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Sigrid Mildenberger
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ralf A. Benndorf
- Department of Clinical Pharmacy and Pharmacotherapy, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Michael Gekle
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
16
|
Pei J, Cong Q. Computational analysis of regulatory regions in human protein kinases. Protein Sci 2023; 32:e4764. [PMID: 37632170 PMCID: PMC10503413 DOI: 10.1002/pro.4764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Eukaryotic proteins often feature modular domain structures comprising globular domains that are connected by linker regions and intrinsically disordered regions that may contain important functional motifs. The intramolecular interactions of globular domains and nonglobular regions can play critical roles in different aspects of protein function. However, studying these interactions and their regulatory roles can be challenging due to the flexibility of nonglobular regions, the long insertions separating interacting modules, and the transient nature of some interactions. Obtaining the experimental structures of multiple domains and functional regions is more difficult than determining the structures of individual globular domains. High-quality structural models generated by AlphaFold offer a unique opportunity to study intramolecular interactions in eukaryotic proteins. In this study, we systematically explored intramolecular interactions between human protein kinase domains (KDs) and potential regulatory regions, including globular domains, N- and C-terminal tails, long insertions, and distal nonglobular regions. Our analysis identified intramolecular interactions between human KDs and 35 different types of globular domains, exhibiting a variety of interaction modes that could contribute to orthosteric or allosteric regulation of kinase activity. We also identified prevalent interactions between human KDs and their flanking regions (N- and C-terminal tails). These interactions exhibit group-specific characteristics and can vary within each specific kinase group. Although long-range interactions between KDs and nonglobular regions are relatively rare, structural details of these interactions offer new insights into the regulation mechanisms of several kinases, such as HASPIN, MAPK7, MAPK15, and SIK1B.
Collapse
Affiliation(s)
- Jimin Pei
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Qian Cong
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|
17
|
Karagöz Z, Passanha FR, Robeerst L, van Griensven M, LaPointe VLS, Carlier A. Computational evidence for multi-layer crosstalk between the cadherin-11 and PDGFR pathways. Sci Rep 2023; 13:15804. [PMID: 37737289 PMCID: PMC10517159 DOI: 10.1038/s41598-023-42624-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: 06/23/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023] Open
Abstract
Various cell surface receptors play an important role in the differentiation and self-renewal of human mesenchymal stem cells (hMSCs). One example of such receptors are the cadherins, which maintain cell-cell adhesion and mechanically couple cells together. Recently, cadherin-11, which is a member of the type II classical cadherin family, has been shown to be involved in the fate commitment of hMSCs. Interestingly, cadherin-11 has no known intrinsic signaling activity and is thought to affect cell behavior via interactions with other cell surface receptors. Members of the platelet-derived growth factor receptor (PDGFR) family are hypothesized to be one of the interaction partners of cadherin-11. Experiments confirmed that PDGFR-α binding to extracellular cadherin-11 regions increases the PDGFR-α activity, whereas the interaction between PDGFR-β and cadherin-11 suppresses the activity of the growth factor receptor. Cadherin-11 knockdown experiments also decreased cell proliferation. These interactions between cadherin-11 and PDGFRs indicate a crosstalk between these receptors and their downstream signaling activities but the nature of this crosstalk is not entirely known. In this study, we used a computational model to represent the experimentally proven interactions between cadherin-11 and the two PDGFRs and we inspected whether the crosstalk also exists downstream of the signaling initiated by the two receptor families. The computational framework allowed us to monitor the relative activity levels of each protein in the network. We performed model simulations to mimic the conditions of previous cadherin-11 knockdown experiments and to predict the effect of crosstalk on cell proliferation. Overall, our predictions suggest the existence of another layer of crosstalk, namely between β-catenin (downstream to cadherin-11) and an ERK inhibitor protein (e.g. DUSP1), different than the crosstalk at the receptor level between cadherin-11 and PDGFR-α and -β. By investigating the multi-level crosstalk between cadherin and PDGFRs computationally, this study contributes to an improved understanding of the effect of cell surface receptors on hMSCs proliferation.
Collapse
Affiliation(s)
- Zeynep Karagöz
- Department of Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Fiona R Passanha
- Department of Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Lars Robeerst
- Department of Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Martijn van Griensven
- Department of Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Vanessa L S LaPointe
- Department of Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|
18
|
Liu Y, Li Y, Tan Q, Lv Y, Tang Y, Yang Y, Yao X, Yang F. Long-Term Exposure to Microcystin-LR Induces Gastric Toxicity by Activating the Mitogen-Activated Protein Kinase Signaling Pathway. Toxins (Basel) 2023; 15:574. [PMID: 37756000 PMCID: PMC10535883 DOI: 10.3390/toxins15090574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Previous studies have primarily concentrated on the hepatotoxicity of MC-LR, whereas its gastric toxicity effects and mechanisms of long-term exposure under low dosage remain unknown. Herein, the gastric tissue from C57BL/6 mice fed with drinking water contaminated by low-dose MC-LR (including 1, 60, and 120 μg/L) was investigated. The results obtained showed that exposure to different concentrations of MC-LR resulted in significant shedding and necrosis of gastric epithelial cells in mice, and a down-regulation of tight junction markers, including ZO-1, Claudin1, and Occludin in the stomach, which might lead to increased permeability of the gastric mucosa. Moreover, the protein expression levels of p-RAF/RAF, p-ERK1/2/ERK1/2, Pink1, Parkin, and LC3-II/LC-3-I were increased in the gastric tissue of mice exposed to 120 μg/L of MC-LR, while the protein expression level of P62 was significantly decreased. Furthermore, we found that pro-inflammatory factors, including IL-6 and TNF-ɑ, were dramatically increased, while the anti-inflammatory factor IL-10 was significantly decreased in the gastric tissue of MC-LR-exposed mice. The activation of the MAPK signaling pathway and mitophagy might contribute to the development of gastric damage by promoting inflammation. We first reported that long-term exposure to MC-LR induced gastric toxicity by activating the MAPK signaling pathway, providing a new insight into the gastric toxic mechanisms caused by MC-LR.
Collapse
Affiliation(s)
- Ying Liu
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China
| | - Yafang Li
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China
| | - Qinmei Tan
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China
| | - Yilin Lv
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China
| | - Yan Tang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China
| | - Yue Yang
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421009, China
| | - Xueqiong Yao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China
| | - Fei Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421009, China
- Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421009, China
| |
Collapse
|
19
|
Anim MT, Tuffour I, Willis R, Schell M, Ostlund T, Mahnashi MH, Halaweish F, Willand-Charnley R. Deacetylated Sialic Acid Sensitizes Lung and Colon Cancers to Novel Cucurbitacin-Inspired Estrone Epidermal Growth Factor Receptor (EGFR) Inhibitor Analogs. Molecules 2023; 28:6257. [PMID: 37687086 PMCID: PMC10488366 DOI: 10.3390/molecules28176257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Cancers utilize sugar residues such as sialic acids (Sia) to improve their ability to survive. Sia presents a variety of functional group alterations, including O-acetylation on the C6 hydroxylated tail. Previously, sialylation has been reported to suppress EGFR activation and increase cancer cell sensitivity to Tyrosine Kinase Inhibitors (TKIs). In this study, we report on the effect of deacetylated Sia on the activity of three novel EGFR-targeting Cucurbitacin-inspired estrone analogs (CIEAs), MMA 294, MMA 321, and MMA 320, in lung and colon cancer cells. Acetylation was modulated by the removal of Sialate O-Acetyltransferase, also known as CAS1 Domain-containing protein (CASD1) gene via CRISPR-Cas9 gene editing. Using a variety of cell-based approaches including MTT cell viability assay, flow cytometry, immunofluorescence assay and in-cell ELISA we observed that deacetylated Sia-expressing knockout cells (1.24-6.49 μM) were highly sensitive to all CIEAs compared with the control cells (8.82-20.97 μM). Apoptosis and varied stage cell cycle arrest (G0/G1 and G2/M) were elucidated as mechanistic modes of action of the CIEAs. Further studies implicated overexpression of CIEAs' cognate protein target, phosphorylated EGFR, in the chemosensitivity of the deacetylated Sia-expressing knockout cells. This observation correlated with significantly decreased levels of key downstream proteins (phosphorylated ERK and mTOR) of the EGFR pathway in knockout cells compared with controls when treated with CIEAs. Collectively, our findings indicate that Sia deacetylation renders lung and colon cancer cells susceptible to EGFR therapeutics and provide insights for future therapeutic interventions.
Collapse
Affiliation(s)
- Mathias T. Anim
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Isaac Tuffour
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Rylan Willis
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Matthew Schell
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Trevor Ostlund
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Mater H. Mahnashi
- Department of Pharmaceutical Chemistry, Najran University, Najran P.O. Box 1988, Saudi Arabia;
| | - Fathi Halaweish
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| | - Rachel Willand-Charnley
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA; (M.T.A.); (I.T.); (R.W.); (M.S.); (T.O.); (F.H.)
| |
Collapse
|
20
|
Yuan M, Lin X, Wang D, Dai J. Proteins: Neglected active ingredients in edible bird's nest. CHINESE HERBAL MEDICINES 2023; 15:383-390. [PMID: 37538855 PMCID: PMC10394320 DOI: 10.1016/j.chmed.2023.02.004] [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: 11/08/2022] [Revised: 01/15/2023] [Accepted: 02/21/2023] [Indexed: 08/05/2023] Open
Abstract
Edible bird's nest (EBN) is a kind of natural invigorant with a long history of consumption in Asia, especially in China. EBN is formed by mixing the saliva of swiftlets (Aerodramus) with feathers and other components during the breeding season. Proteins are the most important nutrient in EBN. By studying proteins in EBN, we can not only elucidate their components at the molecular level, but also study their bioactivities. Therefore, it is of great significance to study the proteins in EBN. Previous research on the proteins in EBN was preliminary and cursory, and no one has summarized and analyzed the proteins in EBN and correlated the bioactivities of these proteins with the biological functions of EBN. This article focused on the proteins in EBN, listed the proteins identified in different proteomic studies, and introduced the sources, structures and bioactivities of the most frequently identified proteins, including acidic mammalian chitinase, lysyl oxidase homolog 3, mucin-5AC, ovoinhibitor, nucleobindin-2, calcium-binding protein (MW: 4.5 × 104) and glucose-regulated protein (MW: 7.8 × 104). The properties of these proteins are closely related to the bioactivities of EBN. Therefore, this article can provide inspiration for further research on the efficacy of EBN.
Collapse
Affiliation(s)
- Man Yuan
- Hebei Edible Bird's Nest Fresh Stew Technology Innovation Center, Langfang 065700, China
| | - Xiaoxian Lin
- Hebei Edible Bird's Nest Fresh Stew Technology Innovation Center, Langfang 065700, China
| | - Dongliang Wang
- Hebei Edible Bird's Nest Fresh Stew Technology Innovation Center, Langfang 065700, China
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jianye Dai
- Hebei Edible Bird's Nest Fresh Stew Technology Innovation Center, Langfang 065700, China
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
21
|
Shi S, Zhang C, Liu J. TIMP2 facilitates CIRI through activating NLRP3-mediated pyroptosis. Aging (Albany NY) 2023; 15:3635-3643. [PMID: 37178321 PMCID: PMC10449283 DOI: 10.18632/aging.204696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023]
Abstract
This study aimed to investigate the underlying mechanisms of cerebral ischemia-reperfusion injury (CIRI) in mice using CIR and hypoxia/reoxygenation (H/R) cell models. The study evaluated brain tissue weight, pathological injury, and changes in the expression levels of TIMP2, p-ERK1/2 and NLRP3-mediated pyroptosis-related proteins in brain tissues and hippocampal neurons of CIR mice using established methods such as dry/wet weight measurement, HE staining, qPCR, TUNEL assay, and Western blotting. The results demonstrated a significant increase in brain water content and neuronal apoptosis rate in the experimental groups compared with those in the control group. In particular, the I/R+TIMP2 group showed the highest increase. Additionally, the control group exhibited a clear brain tissue structure, neatly and densely arranged cells with normal morphology, and evenly stained and clear hippocampal tissues. However, the I/R group showed hippocampal structure disorders, interstitial edema, deep nuclear staining, karyopyknosis, and karyorrhexis in brain tissues. The study results further revealed that TIMP2 could aggravate the pathological damage of brain tissues in the I/R+TIMP2 group compared with the I/R group and significantly reduced it in the TIMP2-KD group. Furthermore, the Western blotting results demonstrated that the protein expression levels of TIMP2, p-ERK1/2, t-ERK1/2, NLRP3, IL-1β, IL-18, GSDMD, Caspase-1, and ASC in brain tissues and hippocampal neurons were significantly higher in the experimental groups than those in the control group. The I/R+TIMP2 group displaying the highest increase and the TIMP2-KD group showing a significant decrease. In conclusion, TIMP2 can contribute to the occurrence and progression of CIRI by activating NLRP3-mediated pyroptosis.
Collapse
Affiliation(s)
- Shaoyong Shi
- Department of Prehospital Emergency Care, Qinhuangdao First Hospital, Qinhuangdao 066000, China
| | - Chongyang Zhang
- Department of Prehospital Emergency Care, Qinhuangdao First Hospital, Qinhuangdao 066000, China
| | - Jiaxiang Liu
- Department of Prehospital Emergency Care, Qinhuangdao First Hospital, Qinhuangdao 066000, China
| |
Collapse
|
22
|
Verzijl CRC, van de Peppel IP, Eilers RE, Bloks VW, Wolters JC, Koehorst M, Kloosterhuis NJ, Havinga R, Jalving M, Struik D, Jonker JW. Pharmacological inhibition of MEK1/2 signaling disrupts bile acid metabolism through loss of Shp and enhanced Cyp7a1 expression. Biomed Pharmacother 2023; 159:114270. [PMID: 36680812 DOI: 10.1016/j.biopha.2023.114270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
The RAS-MAPK signaling pathway is one of the most frequently dysregulated pathways in human cancer. Small molecule inhibitors directed against this pathway have clinical activity in patients with various cancer types and can improve patient outcomes. However, the use of these drugs is associated with adverse effects, which can result in dose reduction or treatment interruption. A better molecular understanding of on-target, off-tumor effects may improve toxicity management. In the present study, we aimed to identify early initiating biological changes in the liver upon pharmacological inhibition of the RAS-MAPK signaling pathway. To this end, we tested the effect of MEK inhibitor PD0325901 using mice and human hepatocyte cell lines. Male C57BL/6 mice were treated with either vehicle or PD0325901 for six days, followed by transcriptome analysis of the liver and phenotypic characterization. Pharmacological MEK inhibition altered the expression of 423 genes, of which 78 were upregulated and 345 were downregulated. We identified Shp, a transcriptional repressor, and Cyp7a1, the rate-limiting enzyme in converting cholesterol to bile acids, as the top differentially expressed genes. PD0325901 treatment also affected other genes involved in bile acid regulation, which was associated with changes in the composition of plasma bile acids and composition and total levels of fecal bile acids and elevated predictive biomarkers of early liver toxicity. In conclusion, short-term pharmacological MEK inhibition results in profound changes in bile acid metabolism, which may explain some of the clinical adverse effects of pharmacological inhibition of the RAS-MAPK pathway, including gastrointestinal complications and hepatotoxicity.
Collapse
Affiliation(s)
- Cristy R C Verzijl
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ivo P van de Peppel
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Roos E Eilers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Justina C Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martijn Koehorst
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9713 Groningen, GZ, The Netherlands
| | - Niels J Kloosterhuis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rick Havinga
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dicky Struik
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johan W Jonker
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
23
|
Wei X, Zhou S, Liao L, Liu M, Gao Y, Yin Y, Xu Q, Zhou R. Comprehensive analysis of transcriptomic profiling of 5-methylcytosin modification in placentas from preeclampsia and normotensive pregnancies. FASEB J 2023; 37:e22751. [PMID: 36692426 DOI: 10.1096/fj.202201248r] [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: 08/03/2022] [Revised: 11/09/2022] [Accepted: 12/21/2022] [Indexed: 01/25/2023]
Abstract
Increasing evidence suggests that RNA m5C modification and its regulators have been confirmed to be associated with the pathogenesis of many diseases. However, the distribution and biological functions of m5C in mRNAs of placental tissues remain unknown. we collected placentae from normotensive pregnancies (CTR) and preeclampsia patients (PE) to analyze the transcriptomic profiling of m5C RNA methylation through m5C RNA immunoprecipitation (UMI-MeRIP-Seq). we discovered that overall m5C methylation peaks were decreased in placental tissues from PE patients. And, 2844 aberrant m5C peaks were identified, of which respectively 1304 m5C peaks were upregulated and 1540 peaks were downregulated. The distribution of m5C peaks were mainly located in CDS (coding sequences) regions in placental tissues of both groups, but compared with the CTR group, the m5C peak in PE group before the stop code of CDS was significantly increased and even higher than the peak value after start code in CDS. Differentially methylated genes were mainly enriched in MAPK/cAMP signaling pathway. Moreover, the up-regulated genes with hypermethylated modification were enriched in the processes of hypoxia, inflammation/immune response. Finally, through analyzing the mRNA expression levels of m5C RNA methylation regulators, we found only DNMT3B and TET3 were significantly upregulated in PE samples than in control group. And they are not only negatively correlated with each other, but also closely related to those differentially expressed genes modified by differential methylation.Our findings provide new insights regarding alterations of m5C RNA modification into the pathogenic mechanisms of PE.
Collapse
Affiliation(s)
- Xiaohong Wei
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, NHC Key Laboratory of Chronobiology, Sichuan University, Ministry of Education, Chengdu, China
| | - Shengping Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, NHC Key Laboratory of Chronobiology, Sichuan University, Ministry of Education, Chengdu, China
| | - Lingyun Liao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, NHC Key Laboratory of Chronobiology, Sichuan University, Ministry of Education, Chengdu, China
| | - Min Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, NHC Key Laboratory of Chronobiology, Sichuan University, Ministry of Education, Chengdu, China
| | - Yijie Gao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, NHC Key Laboratory of Chronobiology, Sichuan University, Ministry of Education, Chengdu, China
| | - Yangxue Yin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, NHC Key Laboratory of Chronobiology, Sichuan University, Ministry of Education, Chengdu, China
| | - Qin Xu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, NHC Key Laboratory of Chronobiology, Sichuan University, Ministry of Education, Chengdu, China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, NHC Key Laboratory of Chronobiology, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
24
|
Wiśniowski T, Bryda J, Wątroba S. The role of matrix metalloproteinases in pathogenesis, diagnostics, and treatment of human prostate cancer. POSTEP HIG MED DOSW 2023. [DOI: 10.2478/ahem-2023-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023] Open
Abstract
Abstract
The prostate gland is highly susceptible to oncogenic transformation, many times more than other sex tissues, such as seminal vesicles. In fact, prostate cancer (PCa) will be diagnosed in one in seven lifetime patients, making PCa the subject of intense research aimed at clarifying its biology and providing adequate treatment. PCa is the fourth most common cancer in the world in terms of the overall population and the second most common cancer for the male population. It is postulated that the development of PCa may be influenced by dietary factors, physical and sexual activity, androgens, obesity, and inflammation, but their role in the development of prostate cancer still remains unclear. Extracellular matrix metalloproteinases (MMPs) and tissue metalloproteinase inhibitors (TIMPs) play an important role in many physiological and pathological processes, including proliferation, migration, invasion, cell differentiation, participation in inflammatory processes and angiogenesis. Numerous studies point to a direct relationship between MMPs and both local tumor invasion and the formation of distant metastases. High activity of MMPs is observed in solid tumors of various origins, which positively correlates with a poor overall survival rate. Although biochemical diagnostic markers of PCa are currently available, from the point of view of clinical practice, it seems particularly important to develop new and more sensitive markers allowing for early diagnosis and long-term monitoring of patients after PCa treatment, and the assessment of MMP activity in urine and serum of patients are potential factors that could play such a role.
Collapse
|
25
|
Cardioprotective Effects of Aconite in Isoproterenol-Induced Myocardial Infarction in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1090893. [PMID: 36600948 PMCID: PMC9807305 DOI: 10.1155/2022/1090893] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 12/27/2022]
Abstract
Background Myocardial infarction (MI) is a severe clinical condition caused by decreased or complete cessation of blood flow to a portion of the myocardium. Aconite, the lateral roots of Aconitum carmichaelii Debx., is a well-known Chinese medicine for treatment of heart failure and related cardiac diseases. The present study is aimed at investigating the cardioprotective effect of aconite on isoproterenol- (ISO)- induced MI. Methods The qualitative analysis of aqueous extracts from brained aconite (AEBA) was conducted by HPLC. A rat model of MI induced by ISO was established to examine the effects of AEBA. The cardiac function was assessed by echocardiography. The serum levels of SOD, CK-MB, cTnT, and cTnI were detected to estimate myocardial injury. The pathological changes of heart tissue were evaluated by 2,3,5-triphenyltetrazolium chloride (TTC) staining, hematoxylin-eosin (HE) staining, and Masson's trichrome staining. The expressions of abnormal vascular remodeling and hypoxia-related components and the levels of inflammation-associated genes and proteins were detected by RT-qPCR, western blotting, and immunofluorescence. Results The contents of benzoylaconine, benzoylmesaconine, benzoylhypacoitine, and hypaconitine in AEBA were 1.35 μg/g, 37.35 μg/g, 57.10 μg/g, and 2.46 μg/g, respectively. AEBA obviously improved heart function through promoting echocardiographic parameters, radial strain, and circumferential strain. The data of TTC staining, HE staining, and Masson's trichrome staining disclosed that AEBA could significantly reduce infarct size, inhibit inflammatory cell infiltration, and decrease the myocardial fibrosis. Moreover, AEBA distinctly suppressed the serum levels of SOD, MDA, CK-MB, cTnT, and cTnI in ISO-induced rats. The results of RT-qPCR indicated that AEBA inhibited the expressions of hypoxia- and inflammation-related genes, including VEGF, PKM2, GLUT-1, LDHA, TNF-α, IL-1β, IL-6, and COX2. In addition, the western blotting and immunofluorescence analyses further confirmed the results of RT-qPCR. Conclusion In summary, our results indicate that the AEBA could improve ISO-induced myocardial infarction by promoting cardiac function, alleviating myocardial hypoxia, and inhibiting inflammatory response and fibrosis in heart tissue.
Collapse
|
26
|
Bon-Mathier AC, Déglise T, Rignault-Clerc S, Bielmann C, Mazzolai L, Rosenblatt-Velin N. Brain Natriuretic Peptide Protects Cardiomyocytes from Apoptosis and Stimulates Their Cell Cycle Re-Entry in Mouse Infarcted Hearts. Cells 2022; 12:cells12010007. [PMID: 36611800 PMCID: PMC9818267 DOI: 10.3390/cells12010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Brain Natriuretic Peptide (BNP) supplementation after infarction increases heart function and decreases heart remodeling. BNP receptors, NPR-A and NPR-B are expressed on adult cardiomyocytes (CMs). We investigated whether a part of the BNP cardioprotective effect in infarcted and unmanipulated hearts is due to modulation of the CM fate. For this purpose, infarcted adult male mice were intraperitoneally injected every two days during 2 weeks with BNP or saline. Mice were sacrificed 1 and 14 days after surgery. BNP or saline was also injected intraperitoneally every two days into neonatal pups (3 days after birth) for 10 days and in unmanipulated 8-week-old male mice for 2 weeks. At sacrifice, CMs were isolated, counted, measured, and characterized by qRT-PCR. The proportion of mononucleated CMs was determined. Immunostainings aimed to detect CM re-entry in the cell cycle were performed on the different hearts. Finally, the signaling pathway activated by BNP treatment was identified in in vitro BNP-treated adult CMs and in CMs isolated from BNP-treated hearts. An increased number of CMs was detected in the hypoxic area of infarcted hearts, and in unmanipulated neonatal and adult hearts after BNP treatment. Accordingly, Troponin T plasma concentration was significantly reduced 1 and 3 days after infarction in BNP-treated mice, demonstrating less CM death. Furthermore, higher number of small, dedifferentiated and mononucleated CMs were identified in adult BNP-treated hearts when compared to saline-treated hearts. BNP-treated CMs express higher levels of mRNAs coding for hif1 alpha and for the different cyclins than CMs isolated from saline-treated hearts. Higher percentages of CMs undergoing DNA synthesis, expressing Ki67, phospho histone3 and Aurora B were detected in all BNP-treated hearts, demonstrating that CMs re-enter into the cell cycle. BNP effect on adult CMs in vivo is mediated by NPR-A binding and activation of the ERK MAP kinase pathway. Interestingly, an increased number of CMs was also detected in adult infarcted hearts treated with LCZ696, an inhibitor of the natriuretic peptide degradation. Altogether, our results identified BNP and all therapies aimed to increase BNP's bioavailability as new cardioprotective targets as BNP treatment leads to an increased number of CMs in neonatal, adult unmanipulated and infarcted hearts.
Collapse
|
27
|
Jin H, Yang B, Jiang D, Ding Z, Xiong Y, Zeng X. Inhibitory effect of anti-Scg3 on corneal neovascularization: a preliminary study. BMC Ophthalmol 2022; 22:455. [PMID: 36443679 PMCID: PMC9703748 DOI: 10.1186/s12886-022-02690-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/16/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Corneal neovascularization (CNV) is an important disease that causes blindness. Secretogranin III (Scg3) has emerged as a new influencing factor of neovascularization. This study analyzed the Scg3 antibody's inhibitory effect on CNV and and explored its preliminary mechanism. METHODS Human umbilical vein endothelial cells (HUVECs) were treated with Scg3 and anti-Scg3. Cell proliferation, wound healing migration and tube formation assays were performed. Healthy adult New Zealand rabbits were randomly selected to be alkali burned and establish the corneal neovascularization (CNV) model. The rabbits were randomly divided into 3 groups (the high concentration group, low concentration group and control group). Different doses of anti-Scg3 and PBS were administered to the rabbits. Clinical examinations, immunostaining, quantitative real-time polymerase chain reaction (qPCR) and western blotting analyses were performed postoperatively. RESULTS In the in vitro study, the Scg3 antibody mixture inhibited Scg3-induced endothelial cell proliferation and angiogenesis. In the in vivo study, significant CNV was observed in the control group. Confocal microscopy also revealed considerable active neovascularization in the control group. There was no obvious CNV growth in the high concentration group. Additionally, CD31, LYVE1 and CD45 expression was significantly inhibited after treatment with a high concentration of Scg3 antibody. The qPCR and western blotting analyses revealed that the levels of ERK in the low concentration group and high concentration group were higher than those in the control group at 7 days and 14 days. The levels of VEGF in the control group were significantly increased compared with those in the high concentration group. In all three groups, the levels of Akt were not significantly different at any time point. CONCLUSION The expression of Scg3 could affect the growth of HUVECs in vitro. Treatment with a high concentration (0.5 µg/mL) of Scg3 antibody reduced the inflammatory response and inhibited the growth of corneal neovascularization after corneal alkali burn injury in rabbits. The MEK/ERK pathway might play an important role in the inhibitory effect of anti-Scg3.
Collapse
Affiliation(s)
- He Jin
- grid.443385.d0000 0004 1798 9548Affiliated Hospital of Guilin Medical University, Guilin Medical University, 541001 Guilin, China
| | - Binbin Yang
- grid.443385.d0000 0004 1798 9548Affiliated Hospital of Guilin Medical University, Guilin Medical University, 541001 Guilin, China
| | - Dongdong Jiang
- grid.443385.d0000 0004 1798 9548Affiliated Hospital of Guilin Medical University, Guilin Medical University, 541001 Guilin, China
| | - Zhixiang Ding
- grid.443385.d0000 0004 1798 9548Affiliated Hospital of Guilin Medical University, Guilin Medical University, 541001 Guilin, China
| | - Yu Xiong
- grid.443385.d0000 0004 1798 9548Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin Medical University, 541001 Guilin, China
| | - Xinsheng Zeng
- grid.443385.d0000 0004 1798 9548Affiliated Hospital of Guilin Medical University, Guilin Medical University, 541001 Guilin, China
| |
Collapse
|
28
|
Poitras T, Zochodne DW. Unleashing Intrinsic Growth Pathways in Regenerating Peripheral Neurons. Int J Mol Sci 2022; 23:13566. [PMID: 36362354 PMCID: PMC9654452 DOI: 10.3390/ijms232113566] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 10/17/2023] Open
Abstract
Common mechanisms of peripheral axon regeneration are recruited following diverse forms of damage to peripheral nerve axons. Whether the injury is traumatic or disease related neuropathy, reconnection of axons to their targets is required to restore function. Supporting peripheral axon regrowth, while not yet available in clinics, might be accomplished from several directions focusing on one or more of the complex stages of regrowth. Direct axon support, with follow on participation of supporting Schwann cells is one approach, emphasized in this review. However alternative approaches might include direct support of Schwann cells that instruct axons to regrow, manipulation of the inflammatory milieu to prevent ongoing bystander axon damage, or use of inflammatory cytokines as growth factors. Axons may be supported by a growing list of growth factors, extending well beyond the classical neurotrophin family. The understanding of growth factor roles continues to expand but their impact experimentally and in humans has faced serious limitations. The downstream signaling pathways that impact neuron growth have been exploited less frequently in regeneration models and rarely in human work, despite their promise and potency. Here we review the major regenerative signaling cascades that are known to influence adult peripheral axon regeneration. Within these pathways there are major checkpoints or roadblocks that normally check unwanted growth, but are an impediment to robust growth after injury. Several molecular roadblocks, overlapping with tumour suppressor systems in oncology, operate at the level of the perikarya. They have impacts on overall neuron plasticity and growth. A second approach targets proteins that largely operate at growth cones. Addressing both sites might offer synergistic benefits to regrowing neurons. This review emphasizes intrinsic aspects of adult peripheral axon regeneration, emphasizing several molecular barriers to regrowth that have been studied in our laboratory.
Collapse
Affiliation(s)
| | - Douglas W. Zochodne
- Neuroscience and Mental Health Institute, Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| |
Collapse
|
29
|
Brancaccio M, Milito A, Viegas CA, Palumbo A, Simes DC, Castellano I. First evidence of dermo-protective activity of marine sulfur-containing histidine compounds. Free Radic Biol Med 2022; 192:224-234. [PMID: 36174879 DOI: 10.1016/j.freeradbiomed.2022.09.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/06/2022] [Accepted: 09/17/2022] [Indexed: 10/31/2022]
Abstract
Among natural products, ovothiol (ovo), produced by marine invertebrates, bacteria, and microalgae, is receiving increasing interest for its unique antioxidant properties. Recently, ovo has been shown to exhibit anti-inflammatory activity in an in vitro model of endothelial dysfunction and in an in vivo model of liver fibrosis. The aim of this study was to evaluate the effect of ovo and its precursor 5-thiohistidine (5-thio) in comparison with ergothioneine (erg), in human skin cells and tissues upon inflammation. We used both an in vitro and ex vivo model of human skin, represented by a keratinocytes cell line (HaCaT) and skin biopsies, respectively. We observed that ovo, 5-thio, and erg were not cytotoxic in HaCaT cells, but instead exerted a protective function against TNF-α -induced inflammation. In order to get insights on their mechanism of action, we performed western blot analysis of ERK and JNK, as well as sub-cellular localization of Nrf2, a key mediator of the anti-inflammatory response. The results indicated that the pre-treatment with ovo, 5-thio, and erg differently affected the phosphorylation of ERK and JNK. However, all the three molecules promoted the accumulation of Nrf2 in the nucleus of HaCaT cells. In addition, gene expression analysis by RTqPCR and ELISA assays performed in ex vivo human skin tissues pre-treated with thiohistidines and then inflamed with IL-1β revealed a significant downregulation of IL-8, TNF-α and COX-2 genes and a concomitant significant decrease in the cytokines IL-6, IL-8 and TNF-α production. Moreover, the protective action of ovo and 5-thio resulted to be stronger when compared with dexamethasone, a corticosteroid drug currently used to treat skin inflammatory conditions. Our findings suggest that ovo and 5-thio can ameliorate skin damage and may be used to develop natural skin care products to prevent the inflammatory status induced by environmental stressors and aging.
Collapse
Affiliation(s)
- Mariarita Brancaccio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
| | - Alfonsina Milito
- Centre for Research in Agricultural Genomics - CRAG, Barcelona, Catalonia, Spain
| | - Carla Alexandra Viegas
- Centre of Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal; GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal
| | - Anna Palumbo
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, Naples, Italy
| | - Dina Costa Simes
- Centre of Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal; GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal
| | - Immacolata Castellano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy; Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, Naples, Italy.
| |
Collapse
|
30
|
Ding Z, Zhao J, Wang X, Li W, Chen C, Yong C, Zhu Y, Tian F, Liu L, Yu M, Zhou E, Gu L, Yao C, Gao K. Total extract of Abelmoschus manihot L. alleviates uric acid-induced renal tubular epithelial injury via inhibition of caspase-8/caspase-3/NLRP3/GSDME signaling. Front Pharmacol 2022; 13:907980. [PMID: 36052125 PMCID: PMC9424722 DOI: 10.3389/fphar.2022.907980] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/08/2022] [Indexed: 11/18/2022] Open
Abstract
Purpose: The incidence of uric acid (UA)-induced kidney injury is increasing owing to the high incidence of hyperuricemia in recent years. The flower of Abelmoschus manihot (Linneus) Medik is a traditional Chinese medicinal herb widely used in the treatment of some kidney diseases. In our previous study, we reported that the total extract of A. manihot L. flower (TEA) attenuated adriamycin-induced renal tubular cell injury. In this study, we aimed to evaluate the role of TEA in UA-induced tubular cell injury. Methods: Normal rat proximal epithelial NRK-52E cells were incubated with UA to mimic hyperuricemia conditions. The role of TEA in the renal tubular cells was also assessed. The cellular morphology was observed using phase-contrast microscopy, and cell viability was analyzed using the Cell Counting kit-8. Living and dead cells were stained using a Calcein-AM/PI double stain kit. The release of lactate dehydrogenase (LDH) was analyzed by LDH cytotoxicity Assay Kit. The expression of target proteins was analyzed using western blot analysis. Results: UA triggered NRK-52E cell injury, as evidenced by morphological changes, detachment of cells from the bottom, cell swelling, large bubbles blowing from cell membrane and loss of cell viability. UA increased release of LDH. UA induced the expression of p-ERK1/2 and the subsequent activation of caspase-8, caspase-3, and NLRP3 inflammasomes. Pyroptosis was elicited by UA after gasdermin E N-terminal (GSDME-NT) was cleaved from gasdermin E (GSDME). Z-DEVD-FMK, a caspase-3 inhibitor, suppressed the expression of both NLRP3 and GSDME-NT, but not that of caspase-8. INF39, an NLRP3 inhibitor, altered the expression of GSDME-NT expression, but not that caspase-3 and caspase-8. TEA alleviated UA-induced cell injury by suppressing ERK1/2/caspase-8/caspase-3/NLRP3/GSDME signaling. Conclusion: GSDME-mediated pyroptosis was involved in UA-induced renal tubular cell injury. This is the first study to report that TEA protects renal tubular epithelial cells against UA by inhibiting the ERK/1/2/caspase-8/caspase-3/NLRP3/GSDME pathway.
Collapse
Affiliation(s)
- Zhihui Ding
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Division of Nephrology, Taizhou Second People’s Hospital, Taizhou, China
| | - Jing Zhao
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xufang Wang
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Li
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chong Chen
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen Yong
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiye Zhu
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Fang Tian
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Liu
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Manshu Yu
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Enchao Zhou
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Liubao Gu
- Division of Clinical Epidemiology, Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Chunlei Yao
- Division of Nephrology, Taizhou Second People’s Hospital, Taizhou, China
- *Correspondence: Kun Gao, ; Chunlei Yao,
| | - Kun Gao
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Kun Gao, ; Chunlei Yao,
| |
Collapse
|
31
|
He L, Zhang CL, Chen Q, Wang L, Huang Y. Endothelial shear stress signal transduction and atherogenesis: From mechanisms to therapeutics. Pharmacol Ther 2022; 235:108152. [PMID: 35122834 DOI: 10.1016/j.pharmthera.2022.108152] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/13/2022] [Accepted: 01/27/2022] [Indexed: 10/19/2022]
Abstract
Atherosclerotic vascular disease and its complications are among the top causes of mortality worldwide. In the vascular lumen, atherosclerotic plaques are not randomly distributed. Instead, they are preferentially localized at the curvature and bifurcations along the arterial tree, where shear stress is low or disturbed. Numerous studies demonstrate that endothelial cell phenotypic change (e.g., inflammation, oxidative stress, endoplasmic reticulum stress, apoptosis, autophagy, endothelial-mesenchymal transition, endothelial permeability, epigenetic regulation, and endothelial metabolic adaptation) induced by oscillatory shear force play a fundamental role in the initiation and progression of atherosclerosis. Mechano-sensors, adaptor proteins, kinases, and transcriptional factors work closely at different layers to transduce the shear stress force from the plasma membrane to the nucleus in endothelial cells, thereby controlling the expression of genes that determine cell fate and phenotype. An in-depth understanding of these mechano-sensitive signaling cascades shall provide new translational strategies for therapeutic intervention of atherosclerotic vascular disease. This review updates the recent advances in endothelial mechano-transduction and its role in the pathogenesis of atherosclerosis, and highlights the perspective of new anti-atherosclerosis therapies through targeting these mechano-regulated signaling molecules.
Collapse
Affiliation(s)
- Lei He
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Cheng-Lin Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen 518060, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Qinghua Chen
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
32
|
Zhang X, Yao Z, Xue Z, Wang S, Liu X, Hu Y, Zhang Y, Wang J, Li X, Chen A. Resibufogenin Targets the ATP1A1 Signaling Cascade to Induce G2/M Phase Arrest and Inhibit Invasion in Glioma. Front Pharmacol 2022; 13:855626. [PMID: 35656311 PMCID: PMC9152115 DOI: 10.3389/fphar.2022.855626] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Resibufogenin (RB) is a major active ingredient in the traditional Chinese medicine Chansu and has garnered considerable attention for its efficacy in the treatment of cancer. However, the anticancer effects and underlying mechanisms of RB on glioblastoma (GBM) remain unknown. Here, we found that RB induced G2/M phase arrest and inhibited invasion in a primary GBM cell line, P3#GBM, and two GBM cell lines, U251 and A172. Subsequently, we demonstrated that RB-induced G2/M phase arrest occurred through downregulation of CDC25C and upregulation of p21, which was caused by activation of the MAPK/ERK pathway, and that RB inhibited GBM invasion by elevating intercellular Ca2+ to suppress the Src/FAK/Paxillin focal adhesion pathway. Intriguingly, we confirmed that upon RB binding to ATP1A1, Na+-K+-ATPase was activated as a receptor and then triggered the intracellular MAPK/ERK pathway and Ca2+-mediated Src/FAK/Paxillin focal adhesion pathway, which led to G2/M phase arrest and inhibited the invasion of GBM cells. Taken together, our findings reveal the antitumor mechanism of RB by targeting the ATP1A1 signaling cascade and two key signaling pathways and highlight the potential of RB as a new class of promising anticancer agents.
Collapse
Affiliation(s)
- Xun Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling and Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Zhong Yao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling and Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Zhiyi Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling and Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Shuai Wang
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Xuemeng Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling and Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Yaotian Hu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling and Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Yan Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling and Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling and Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling and Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling and Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| |
Collapse
|
33
|
Liu Z, Guan C, Li C, Zhang N, Yang C, Xu L, Zhou B, Zhao L, Luan H, Man X, Xu Y. Tilianin Reduces Apoptosis via the ERK/EGR1/BCL2L1 Pathway in Ischemia/Reperfusion-Induced Acute Kidney Injury Mice. Front Pharmacol 2022; 13:862584. [PMID: 35721209 PMCID: PMC9204490 DOI: 10.3389/fphar.2022.862584] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Acute kidney injury (AKI) is a common syndrome impacting about 13.3 million patients per year. Tilianin has been reported to alleviate myocardial ischemia/reperfusion (I/R) injury, while its effect on AKI is unknown; thus, this study aimed to explore if tilianin protects I/R-induced AKI and the underlying mechanisms.Methods: The microarray dataset GSE52004 was downloaded from GEO DataSets (Gene Expression Omnibus). Differential expression analysis and gene-set enrichment analysis (GSEA) were performed by R software to identify apoptosis pathway-related genes. Then, RcisTarget was applied to identify the transcription factor (TF) related to apoptosis. The STRING database was used to construct a protein–protein interaction (PPI) network. Cytoscape software visualized PPI networks, and hub TFs were selected via cytoHubba. AutoDock was used for molecular docking of tilianin and hub gene-encoded proteins. The expression levels of hub genes were assayed and visualized by quantitative real-time PCR, Western blotting, and immunohistochemistry by establishing I/R-induced AKI mouse models.Results: Bioinformatics analysis showed that 34 genes, including FOS, ATF4, and Gadd45g, were involved in the apoptosis pathway. In total, seven hub TFs might play important roles in tilianin-regulating apoptosis pathways. In in vivo, tilianin improved kidney function and reduced the number of TUNEL-positive renal tubular epithelial cells (RTECs) after I/R-induced AKI. Tilianin reduced the activation of the ERK pathway and then downregulated the expression of EGR1. This further ameliorated the expression of anti-apoptotic genes such as BCL2L1 and BCL2, reduced pro-apoptotic genes such as BAD, BAX, and caspase-3, and reduced the release of cytochrome c.Conclusion: Tilianin reduced apoptosis after I/R-induced AKI by the ERK/EGR1/BCL2L1 pathway. Our findings provided novel insights for the first time into the protective effect and underlying molecular mechanisms of tilianin on I/R-induced AKI.
Collapse
Affiliation(s)
- Zengying Liu
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chen Guan
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chenyu Li
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, München, Germany
| | - Ningxin Zhang
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chengyu Yang
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lingyu Xu
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Zhou
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Long Zhao
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Luan
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaofei Man
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Xu
- Department of Nephrology, the Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Yan Xu,
| |
Collapse
|
34
|
Kim B, Breton S. The MAPK/ERK signaling pathway regulates the expression and localization of Cx43 in mouse proximal epididymis†. Biol Reprod 2022; 106:919-927. [PMID: 35156117 PMCID: PMC9113436 DOI: 10.1093/biolre/ioac034] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/17/2022] [Accepted: 02/09/2022] [Indexed: 01/25/2023] Open
Abstract
This study aimed to clarify the functional role of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK1/2)-signaling pathway in the expression and localization of connexin 43 (Cx43). Mice were treated with the mitogen-activated protein kinase kinase (MEK1/2) inhibitor, PD325901, which induced a progressive decrease in ERK1/2 phosphorylation (pERK) in the proximal epididymis of the mice, without affecting total ERK level. Cx43 staining with punctuated reactive sites was observed in the basolateral membranes in the initial segment (IS) of mouse epididymis. However, PD325901 induced a significant decrease in Cx43 labeling in the basolateral membranes. Interestingly, Cx43, which was undetectable in the apical region of epididymis under control conditions, showed a significant increase in the apical region after PD 325901 treatment. To confirm whether Cx43 was present in tight junctions (TJs) after PD 325901 treatment, PD325901-treated epididymis samples were double-labeled with Cx43 and zonula occludens (ZO)-1 (a TJ protein marker). Thereafter, confocal microscopy showed the colocalization of Cx43 and ZO-1 in the epididymis after PD325901 treatment. Collectively, our results indicated that PD325901 treatment induced a significant increase in Cx43 localization on TJs, where it was colocalized with ZO-1. Therefore, the study suggested that ERK phosphorylation is essential for the proper expression and localization of the gap junction (GJ) protein, and that the relationship between GJs and TJs could play an important role in establishing and maintaining microenvironmental homeostasis for sperm maturation in the IS of mouse epididymis.
Collapse
Affiliation(s)
- Bongki Kim
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Animal Resources Science, Kongju National University, Yesan, Chungcheongnam-do, Republic of Korea
| | - Sylvie Breton
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Qi Y, Zhao T, Li R, Han M. Macrophage-Secreted S100A4 Supports Breast Cancer Metastasis by Remodeling the Extracellular Matrix in the Premetastatic Niche. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9895504. [PMID: 35496059 PMCID: PMC9046007 DOI: 10.1155/2022/9895504] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/14/2021] [Accepted: 03/25/2022] [Indexed: 12/03/2022]
Abstract
Metastasis is the major cause of cancer-related mortalities. A tumor-supportive microenvironment, also known as the premetastatic niche at secondary tumor sites, plays a crucial role in metastasis. Remodeling of the extracellular matrix (ECM) is essential for premetastatic niche formation, especially for circulating tumor cell colonization. However, the underlying molecular mechanism that contributes to this effect remains unclear. Here, we developed a lung metastasis model with 4T1 breast cancer cells and found that the metastasis critically depended on the early recruitment of macrophages to the lung. Disruption of macrophage recruitment reduced fibroblast activation and lung metastasis. Furthermore, we identified the secreted protein S100A4, which is produced by M2 macrophages and participates in fibroblast activation and ECM protein deposition via the ERK signaling pathway. Collectively, these results indicate that recruiting S100A4-expressing inflammatory macrophages plays a vital role in ECM remodeling in the premetastatic niche and may act as a potential therapeutic target for breast cancer lung metastasis.
Collapse
Affiliation(s)
- Yana Qi
- Cancer Therapy and Research Center, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021 Shandong, China
| | - Tingting Zhao
- Cancer Therapy and Research Center, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021 Shandong, China
| | - Ranran Li
- Cancer Therapy and Research Center, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021 Shandong, China
| | - Mingyong Han
- Cancer Therapy and Research Center, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021 Shandong, China
| |
Collapse
|
36
|
ROS-responsive thioketal-linked alginate/chitosan carriers for irritable bowel syndrome with diarrhea therapy. Int J Biol Macromol 2022; 209:70-82. [PMID: 35351547 DOI: 10.1016/j.ijbiomac.2022.03.118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/09/2022] [Accepted: 03/17/2022] [Indexed: 02/08/2023]
Abstract
A colon-specific carrier that can protect drugs from the destruction in the gastrointestinal tract is critical for treating irritable bowel syndrome with diarrhea (IBS-D). In this study, chitosan was cross-linked by the thioketal (TK) bond to serve as a ROS-sensitive core of microspheres. Then the chitosan core was coated with an alginate shell. The alginate/chitosan microspheres can protect puerarin against the destruction and elimination in the gastrointestinal tract and release puerarin at the lesion sites in large quantities. The microspheres were characterized using differential scanning calorimetry, Fourier-transform infrared spectroscopy, and scanning electron microscopy. The swelling study showed that microspheres would shrink in an acidic environment. The in vitro release analysis indicated that little puerarin was released at gastric pH but burst release was observed in simulated colonic fluid containing H2O2. Fluorescent tracer revealed that the fluorescence of microspheres lasted up to 30 h in the colon, which was beneficial to prolong the action time between puerarin and colon. The in vivo studies indicated that puerarin-loaded microspheres are more effective in the treatment of IBS-D than free puerarin. Altogether, the ROS-responsive alginate/chitosan microspheres may be a promising strategy for IBS-D.
Collapse
|
37
|
Wang Z, Franke K, Bal G, Li Z, Zuberbier T, Babina M. MRGPRX2-Mediated Degranulation of Human Skin Mast Cells Requires the Operation of Gαi, Gαq, Ca++ Channels, ERK1/2 and PI3K—Interconnection between Early and Late Signaling. Cells 2022; 11:cells11060953. [PMID: 35326404 PMCID: PMC8946553 DOI: 10.3390/cells11060953] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 02/07/2023] Open
Abstract
The recent discovery of MRGPRX2 explains mast cell (MC)-dependent symptoms independently of FcεRI-activation. Because of its novelty, signaling cascades triggered by MRGPRX2 are rudimentarily understood, especially in cutaneous MCs, by which MRGPRX2 is chiefly expressed. Here, MCs purified from human skin were used following preculture or ex vivo and stimulated by FcεRI-aggregation or MRGPRX2 agonists (compound 48/80, Substance P) in the presence/absence of inhibitors. Degranulation was assessed by β-hexosaminidase or histamine release. Phosphorylation events were studied by immunoblotting. As a G protein-coupled receptor, MRGPRX2 signals by activating G proteins; however, their nature has remained controversial. In skin MCs, Gαi and Gαq were required for degranulation, but Gαi was clearly more relevant. Ca++ channels were likewise crucial. Downstream, PI3K was essential for granule discharge initiated by MRGPRX2 or FcεRI. ERK1/2 and JNK were additional participants, especially in the allergic route. Addressing possible points of intersection between early and later events, pERK1/2 and pAKT were found to depend on Gαi, further highlighting its significance. Gαq and Ca++ channels made some contributions to the phosphorylation of ERK. Ca++ differentially affected PI3K activation in FcεRI- vis-à-vis MRGPRX2-signaling, as channel inhibition increased pAKT only when triggered via FcεRI. Collectively, our study significantly extends our understanding of the molecular framework behind granule secretion from skin MCs.
Collapse
Affiliation(s)
- Zhao Wang
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Department of Dermatology, The Second Affiliated Hospital, Northwest Hospital, Xi’an Jiaotong University, Xi’an 710004, China
| | - Kristin Franke
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Gürkan Bal
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Zhuoran Li
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Torsten Zuberbier
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Magda Babina
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
- Correspondence:
| |
Collapse
|
38
|
Zarei S, Taghian F, Sharifi G, Abedi H. Novel prevention insights into depletion of oxidative stress status through regular exercise and grape seed effective substance in heart ischemia rat model. Food Sci Nutr 2022; 10:833-845. [PMID: 35311161 PMCID: PMC8907746 DOI: 10.1002/fsn3.2714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 01/02/2023] Open
Abstract
Myocardial ischemia (MI) is recognized as the most frequent cardiovascular disease which is the dominant cause of global morbidity and mortality. Artificial intelligence tools and integrative data analysis revealed superoxide dismutase, catalase, glutathione peroxidase, gap junction protein α, myosin heavy chains, and zinc finger transcription factor GATA4 are engaged in oxidative stress and in cardiomyopathy. Network analysis indicated that MAPK3 might be the highest distribution property and cut point in this network, which could be a potential candidate for preventing and treating oxidative stress in heart tissue. Among antioxidant agents, grape seed extract (GSE) is an effective substance that altered antioxidant status in heart tissue. Considering drug discovery methods, we illustrated that GSE might target the MAPK3 protein with sufficient binding affinity. Moreover, we found that low- and moderate-intensity training might prevent the depletion of antioxidants after MI. GSE consumption altered the levels of superoxide dismutase, glutathione peroxidase, and catalase after 14 weeks. Therefore, the interaction of low- and moderate-intensity training and GSE had a synergistic effect on the antioxidant status and relative expression of the Mapk3. Moreover, the interaction of high-intensity training and GSE had a compensatory mechanism that could scavenge reactive oxygen species and improve endogenous antioxidants and modulate the Mapk3 level in MI rats. Consequently, we displayed positive influence and synergic effects of simultaneous GSE prescription and regular physical activity for 14 weeks to prevent acute and chronic heart ischemia cardioprotective phenomenon. Furthermore, the capacitation oxidative stress and relative expression of the Mapk3 was significantly increased by GSE and regular exercise.
Collapse
Affiliation(s)
- Safar Zarei
- Department of Sports PhysiologyFaculty of sports sciencesIsfahan (Khorasgan) BranchIslamic Azad UniversityIsfahanIran
| | - Farzaneh Taghian
- Department of Sports PhysiologyFaculty of sports sciencesIsfahan (Khorasgan) BranchIslamic Azad UniversityIsfahanIran
| | - Gholamreza Sharifi
- Department of Sports PhysiologyFaculty of sports sciencesIsfahan (Khorasgan) BranchIslamic Azad UniversityIsfahanIran
| | - Hassanali Abedi
- Research Center for Noncommunicable DiseasesFaculty of MedicineJahrom University of Medical SciencesJahromIran
| |
Collapse
|
39
|
Song Y, Yang X, Zhang X, Zhu J, Chen Y, Gao F, Zhang H, Han Y, Weng Q, Yuan Z. Seasonal expression of extracellular signal regulated kinases in the colon of wild ground squirrels (Spermophilus dauricus). Mol Biol Rep 2022; 49:2209-2215. [PMID: 35040005 DOI: 10.1007/s11033-021-07042-0] [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: 08/23/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND The purpose of the experiment was to explore the localization and seasonal expression of extracellular signal regulated kinase (ERK) in the colonic tissue of wild ground squirrels (Spermophilus dauricus). METHODS AND RESULTS Hematoxylin-eosin staining, immunohistochemistry, real-time quantitative PCR and Western blotting were used in this experiment. The histological results showed that the diameter of the colon lumen enlarged and the number of glandular cells increased in the non-breeding season. It was found in the immunochemical results that both ERK1/2 and pERK1/2 were expressed in the cytoplasm of goblet cells and intestinal epithelial cells, while pERK1/2 was also expressed in the nucleus of them. The immune localization of both was more obvious in the non-breeding season, especially in intestinal epithelial cells. Real-time quantitative PCR and Western blotting showed that ERK1/2 and pERK1/2 were seasonally highly expressed in the non-breeding season. CONCLUSIONS The expression of ERK1/2 and pERK1/2 was seasonal changes and had significant increases in the non-breeding season. This study revealed that ERK1/2 had potential roles in the colon to the adaptation of seasonal changes in wild ground squirrels.
Collapse
Affiliation(s)
- Yue Song
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Xiaoying Yang
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Xueying Zhang
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Jueyu Zhu
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Yixin Chen
- School of Information Science and Technology, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Fuli Gao
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Haolin Zhang
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Yingying Han
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Qiang Weng
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, 100083, People's Republic of China
| | - Zhengrong Yuan
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, 100083, People's Republic of China.
| |
Collapse
|
40
|
Li M, Zhao X, Yong H, Shang B, Lou W, Wang Y, Bai J. FBXO22 Promotes Growth and Metastasis and Inhibits Autophagy in Epithelial Ovarian Cancers via the MAPK/ERK Pathway. Front Pharmacol 2021; 12:778698. [PMID: 34950036 PMCID: PMC8688818 DOI: 10.3389/fphar.2021.778698] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
E3 ubiquitin ligase F-box only protein 22 (FBXO22), which targets the key regulators of cellular activities for ubiquitylation and degradation, plays an important role in tumorigenesis and metastasis. However, the function of FBXO22 in epithelial ovarian cancers has not been reported. This study aims to explore the biological function of FBXO22 in epithelial ovarian cancers progression and metastasis and its specific regulation mechanism. Immunohistochemistry analysis of tissue microarray was performed to evaluate the expression of FBXO22 in epithelial ovarian cancers patients. The proliferative ability of epithelial ovarian cancers cells was examined by the CCK8. The metastasis ability was detected by the wound healing assay, migration and invasion assays. Western blot was used to verify the relationship between FBXO22 expression and mitogen-activated protein kinase related proteins. Autophagic flux was detected by electron microscopy, mRFP-GFP-LC3 adenovirus, lysosomal tracker and western blot. For in vivo experiments, the effect of FBXO22 on epithelial ovarian cancers resistance was observed in a xenograft tumor model and a metastatic mice model. We found that FBXO22 expression was significantly increased in epithelial ovarian cancers tissues and was closely correlated with clinical pathological factors. As a result, we found that FBXO22 promoted the growth and metastasis, as well as inhibited the autophagy flux. In addition, we identified that FBXO22 performed these functions via the MAPK/ERK pathway. Our results first reported the function of FBXO22 in epithelial ovarian cancer and the correlation between FBXO22 and autophagy, suggesting FBXO22 as a novel target of epithelial ovarian cancers assessment and treatment.
Collapse
Affiliation(s)
- Minle Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Xue Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Hongmei Yong
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| | - Bingqing Shang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Weihua Lou
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - You Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
41
|
Sun Q, Zhou Y, Xiong M, Chen Y, Tan WS, Cai H. MEK1 activation enhances the ex vivo proliferation of haematopoietic stem/progenitor cell. Cell Biochem Funct 2021; 40:79-89. [PMID: 34855220 DOI: 10.1002/cbf.3677] [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: 07/06/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 11/11/2022]
Abstract
Haematopoietic stem/progenitor cell (HSPC) integrates intracellular signal network from growth factors (GFs) and utilizes its proliferation feature to generate high yields of transplantable cells upon ex vivo culture. However, the molecular basis for HSPC activation and proliferation is not completely understood. The goal of this study was to investigate proliferation regulator in the downstream of GFs and develop HSPC expansion strategy. Microarray and Ingenuity Pathway Analysis were performed to evaluate differentially expressed genes in cytokine-induced CD34+ cells after ex vivo culture. We identified that MEK1 was a potential HSPC proliferation regulator, which represented indispensable roles and MEK1 silence attenuated the proliferation of HSPC. Notably, 500 nM MEK1 agonist, PAF C-16, increased the numbers of phenotypic HSPC and induced cell cycling of HSPC. The PAF C-16 expanded HSPC demonstrated comparative clonal formation ability and secondary expansion capacity compared to the vehicle control. Our results provide insights into regulating the balance between proliferation and commitment of HSPC by targeting the HSPC proliferation-controlling network. This study demonstrates that MEK1 critically regulates HSPC proliferation and cell production in the ex vivo condition for transplantation.
Collapse
Affiliation(s)
- Qihao Sun
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yiran Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Minghao Xiong
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yuying Chen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
42
|
Mayfosh AJ, Nguyen TK, Hulett MD. The Heparanase Regulatory Network in Health and Disease. Int J Mol Sci 2021; 22:11096. [PMID: 34681753 PMCID: PMC8541136 DOI: 10.3390/ijms222011096] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 12/24/2022] Open
Abstract
The extracellular matrix (ECM) is a structural framework that has many important physiological functions which include maintaining tissue structure and integrity, serving as a barrier to invading pathogens, and acting as a reservoir for bioactive molecules. This cellular scaffold is made up of various types of macromolecules including heparan sulfate proteoglycans (HSPGs). HSPGs comprise a protein core linked to the complex glycosaminoglycan heparan sulfate (HS), the remodeling of which is important for many physiological processes such as wound healing as well as pathological processes including cancer metastasis. Turnover of HS is tightly regulated by a single enzyme capable of cleaving HS side chains: heparanase. Heparanase upregulation has been identified in many inflammatory diseases including atherosclerosis, fibrosis, and cancer, where it has been shown to play multiple roles in processes such as epithelial-mesenchymal transition, angiogenesis, and cancer metastasis. Heparanase expression and activity are tightly regulated. Understanding the regulation of heparanase and its downstream targets is attractive for the development of treatments for these diseases. This review provides a comprehensive overview of the regulators of heparanase as well as the enzyme's downstream gene and protein targets, and implications for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Alyce J. Mayfosh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia; (A.J.M.); (T.K.N.)
| | - Tien K. Nguyen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia; (A.J.M.); (T.K.N.)
| | - Mark D. Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia; (A.J.M.); (T.K.N.)
| |
Collapse
|
43
|
Caliva MJ, Yang WS, Young-Robbins S, Zhou M, Yoon H, Matter ML, Grimes ML, Conrads T, Ramos JW. Proteomics analysis identifies PEA-15 as an endosomal phosphoprotein that regulates α5β1 integrin endocytosis. Sci Rep 2021; 11:19830. [PMID: 34615962 PMCID: PMC8494857 DOI: 10.1038/s41598-021-99348-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022] Open
Abstract
Endosomal trafficking of cell surface receptors is essential to their function. Integrins are transmembrane receptors that integrate adhesion to the extracellular matrix with engagement of the cytoskeleton. Ligated integrins mediate diverse signals that regulate matrix assembly, cell survival, cell morphology, and cell motility. Endosomal trafficking of integrins modulates these signals and contributes to cell motility and is required for cancer cell invasion. The phosphoprotein PEA-15 modulates integrin activation and ERK MAP Kinase signaling. To elucidate novel PEA-15 functions we utilized an unbiased proteomics approach. We identified several binding partners for PEA-15 in the endosome including clathrin and AP-2 as well as integrin β1 and other focal adhesion complex proteins. We confirmed these interactions using proximity ligation analysis, immunofluorescence imaging, pull-down and co-immunoprecipitation. We further found that PEA-15 is enriched in endosomes and was required for efficient endosomal internalization of α5β1 integrin and cellular migration. Importantly, PEA-15 promotion of migration was dependent on PEA-15 phosphorylation at serines 104 and 116. These data support a novel endosomal role for PEA-15 in control of endosomal trafficking of integrins through an association with the β1 integrin and clathrin complexes, and thereby regulation of cell motility.
Collapse
Affiliation(s)
- Maisel J Caliva
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Mānoa, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Won Seok Yang
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Mānoa, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Shirley Young-Robbins
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Mānoa, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Ming Zhou
- Women's Health Integrated Research Center at Inova, Inova Women's Service Line, Inova Health System, 3289 Woodburn Rd, Suite 375, Falls Church, VA, 22003, USA
| | - Hana Yoon
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Mānoa, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Michelle L Matter
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Mānoa, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Mark L Grimes
- Division of Biological Sciences, Center for Structural and Functional Neuroscience, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
| | - Thomas Conrads
- Women's Health Integrated Research Center at Inova, Inova Women's Service Line, Inova Health System, 3289 Woodburn Rd, Suite 375, Falls Church, VA, 22003, USA
| | - Joe William Ramos
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Mānoa, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| |
Collapse
|
44
|
Yang H, Xu Z, Peng Y, Wang J, Xiang Y. Integrin β4 as a Potential Diagnostic and Therapeutic Tumor Marker. Biomolecules 2021; 11:biom11081197. [PMID: 34439865 PMCID: PMC8394641 DOI: 10.3390/biom11081197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 11/05/2022] Open
Abstract
Integrin β4 (ITGβ4) is a class of transmembrane adhesion molecules composed of hemidesmosomes (HDs). Its unique long intracellular domain provides intricate signal transduction functions. These signal transduction effects are especially prominent in tumors. Many recent studies have shown that integrin β4 is differentially expressed in various tumors, and it plays a vital role in tumor invasion, proliferation, epithelial–mesenchymal transition, and angiogenesis. Therefore, we categorize the research related to integrin β4, starting from its structure and function in tumor tissues, and provide a basic description. Based on its structure and function, we believe that integrin β4 can be used as a tumor marker. In clinical practice, it is described as a diagnostic marker for the targeted treatment of cancer and will be helpful in the clinical diagnosis and treatment of tumors.
Collapse
Affiliation(s)
- Haoyu Yang
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Zixuan Xu
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Yuqian Peng
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Jiali Wang
- Xiang Ya School of Medicine, Central South University, Changsha 410013, China;
| | - Yang Xiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Correspondence: ; Tel.:+86-139-7312-8943
| |
Collapse
|
45
|
Recent developments in mitogen activated protein kinase inhibitors as potential anticancer agents. Bioorg Chem 2021; 114:105161. [PMID: 34328852 DOI: 10.1016/j.bioorg.2021.105161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 01/06/2023]
Abstract
The mitogen activated protein kinase (MAPK) belongs to group of kinase that links the extracellular stimuli to intracellular response. The MAPK signalling pathway (RAS-RAF-MEK-ERK) involved in different pathological conditions like cancer, caused due to genetic or any other factor such as physical or environmental. Many studies have been conducted on the pathological view of MAPK cascade and its associated element like RAS, RAF, MEK, ERK or its isoforms, and still the research is going on particularly with respect to its activation, regulation and inhibition. The MAPK signalling pathway has become the area of research to identify new target for the management of cancer. A number of heterocyclics are key to fight with the cancer associated with these enzymes thus give some hope in the management of cancer by inhibiting MAPK cascade. In the present article, we have focussed on MAPK signalling pathway and role of different heterocyclic scaffolds bearing nitrogen, sulphur and oxygen and about their potential to block MAPK signalling pathway. The heterocyclics are gaining importance due to high potency and selectivity with less off-target effects against different targets involved in the MAPK signalling pathway. We have tried to cover recent advancements in the MAPK signalling pathway inhibitors with an aim to get better understanding of the mechanism of action of the compounds. Several compounds in the preclinical and clinical studies have been thoroughly dealt with. In addition to the synthetic compounds, a significant number of natural products containing heterocyclic moieties as MAPK signalling pathway inhibitors have been put together. The structure activity relationship along with docking studies have been discussed to apprehend the mechanistic studies of various compounds that will ultimately help to design and develop more MAPK signalling pathway inhibitors.
Collapse
|
46
|
Miquelianin Inhibits Allergic Responses in Mice by Suppressing CD4 + T Cell Proliferation. Antioxidants (Basel) 2021; 10:antiox10071120. [PMID: 34356353 PMCID: PMC8301087 DOI: 10.3390/antiox10071120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Allergic diseases, including atopic dermatitis (AD), induce type 2 helper T (Th2) cell-dominant immune responses. Miquelianin (quercetin 3-O-glucuronide, MQL) is an active compound in Rosae multiflorae fructus extract with anti-allergic properties. Here, we investigate the anti-allergic effects of MQL in an ovalbumin (OVA)-induced Th2-dominant mouse model and the associated mechanisms. Oral MQL suppressed cytokine and IL-2 production and proliferation of Th2 cells and upregulated heme oxygenase-1 (HO-1) in splenocytes. Ex vivo MQL suppressed Th1- and Th2-related immune responses by inhibiting CD4+ T cell proliferation, and upregulated HO-1 in CD4+ T cells by activating C-Raf-ERK1/2-Nrf2 pathway via induction of reactive oxygen species generation. In a trimellitic anhydride-induced AD-like mouse model, both topical and oral MQL ameliorated AD symptoms by suppressing Th2 immune responses. Our results suggest that MQL is a potential therapeutic agent for CD4+ T cell-mediated diseases, including allergic diseases.
Collapse
|
47
|
Meng X, Chen Y, Macip S, Leppard K. PML-II regulates ERK and AKT signal activation and IFNα-induced cell death. Cell Commun Signal 2021; 19:70. [PMID: 34215258 PMCID: PMC8252201 DOI: 10.1186/s12964-021-00756-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/03/2021] [Indexed: 11/23/2022] Open
Abstract
Background The requirement of promyelocytic leukaemia protein (PML) in interferon (IFN)-induced cell apoptosis is well-established. However, the exact mechanisms by which the multiple isoforms of PML protein participate in this process remain not well-understood. We previously demonstrated that PML isoform II (PML-II) positively regulates induced gene expression during a type I IFN response and evaluate here how PML-II contributes to IFNα-induced cell death. Methods HeLa cells were transiently depleted of PML-II by siRNA treatment and the response of these cells to treatment with IFNα assessed by molecular assays of mRNA and proteins associated with IFN and apoptosis responses. Results In HeLa cells, death during IFNα stimulation was reduced by prior PML-II depletion. PML-II removal also considerably decreased the induced expression of pro-apoptotic ISGs such as ISG54 (IFIT2), and substantially impaired or prevented expression of PUMA and TRAIL, proteins that are associated with the intrinsic and extrinsic apoptotic pathways respectively. Thirdly, PML-II depletion enhanced ERK and AKT pro-survival signaling activation suggesting that PML-II normally suppresses signaling via these pathways, and that lack of PML-II hence led to greater than normal activation of AKT signaling upon IFNα stimulation and consequently increased resistance to IFNα-induced apoptosis. Conclusions The positive contribution of PML-II to the expression of various IFNα-induced pro-apoptotic proteins and its inhibition of pro-survival signaling together provide a mechanistic explanation for reduced apoptosis under conditions of PML deficiency and may account for at least part of the role of PML as a tumor suppressor gene. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-021-00756-5.
Collapse
Affiliation(s)
- Xueqiong Meng
- School of Basic Medicine, Henan University of Science and Technology, Luoyang, China.,School of Life Sciences, University of Warwick, Coventry, UK
| | - Yixiang Chen
- School of Basic Medicine, Henan University of Science and Technology, Luoyang, China.,Henan International Joint Laboratory of Thrombosis and Hemostasis, Luoyang, China.,Mechanisms of Cancer and Aging Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Salvador Macip
- Mechanisms of Cancer and Aging Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK.,FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Keith Leppard
- School of Life Sciences, University of Warwick, Coventry, UK.
| |
Collapse
|
48
|
Verschoor PJ, Greig FH, Rochford JJ, Levate G, Delibegovic M, Thompson D, Leeson-Payne A, Dekeryte R, Banks R, Ramos JW, Nixon GF. Phosphoprotein enriched in astrocytes (PEA)-15 is a novel regulator of adipose tissue expansion. Sci Rep 2021; 11:6949. [PMID: 33772049 PMCID: PMC7997924 DOI: 10.1038/s41598-021-86250-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/26/2021] [Indexed: 12/17/2022] Open
Abstract
Excessive expansion of adipose tissue in obesity typically leads to overflow and accumulation of lipids in other tissues, causing fatty liver disease and atherosclerosis. The intracellular protein, phosphoprotein enriched in astrocytes (PEA)-15 has been linked to metabolic disease but its role in lipid storage has not been examined. To delineate the role of PEA-15 in adipose tissue, we placed PEA-15−/− mice on a high fat diet. These mice developed increased body weight and greater white adipose tissue expansion compared to high fat diet-fed wild type mice. This was due to increased adipocyte cell size in PEA-15−/− mice consistent with greater lipid storage capacity. Surprisingly, PEA-15−/− mice exhibited improvements in whole body insulin sensitivity, lower hepatic weight and decreased serum triglycerides indicating a protective phenotype. To determine effects on atherosclerosis, PEA-15−/− mice were crossed with the ApoE−/− mice on a high fat diet. Strikingly, these mice were protected from atherosclerosis and had less hepatic lipid accumulation despite increased adiposity. Therefore, we reveal for the first time that PEA-15 plays a novel role in regulating the expansion of adipose tissue. Decreasing PEA-15 expression increases the sequestering of lipids in adipose tissue, protecting other tissues in obesity, thereby improving metabolic health.
Collapse
Affiliation(s)
- Pola J Verschoor
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Fiona H Greig
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Justin J Rochford
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.,Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Giovanni Levate
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Mirela Delibegovic
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Dawn Thompson
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | | | - Ruta Dekeryte
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Ruth Banks
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Joe W Ramos
- University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, USA
| | - Graeme F Nixon
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
49
|
Barbosa R, Acevedo LA, Marmorstein R. The MEK/ERK Network as a Therapeutic Target in Human Cancer. Mol Cancer Res 2021; 19:361-374. [PMID: 33139506 PMCID: PMC7925338 DOI: 10.1158/1541-7786.mcr-20-0687] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/01/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022]
Abstract
The RAS-RAF-MEK-ERK pathway is the most well-studied of the MAPK cascades and is critical for cell proliferation, differentiation, and survival. Abnormalities in regulation resulting from mutations in components of this pathway, particularly in upstream proteins, RAS and RAF, are responsible for a significant fraction of human cancers and nearly all cutaneous melanomas. Activation of receptor tyrosine kinases by growth factors and various extracellular signals leads to the sequential activation of RAS, RAF, MEK, and finally ERK, which activates numerous transcription factors and facilitates oncogenesis in the case of aberrant pathway activation. While extensive studies have worked to elucidate the activation mechanisms and structural components of upstream MAPK components, comparatively less attention has been directed toward the kinases, MEK and ERK, due to the infrequency of oncogenic-activating mutations in these kinases. However, acquired drug resistance has become a major issue in the treatment of RAS- and RAF-mutated cancers. Targeting the terminal kinases in the MAPK cascade has shown promise for overcoming many of these resistance mechanisms and improving treatment options for patients with MAPK-aberrant cancers. Here, we will describe the role of MEK and ERK in MAPK signaling and summarize the current understanding of their interaction and activation mechanisms. We will also discuss existing approaches for targeting MEK and ERK, and the benefits of alternative strategies. Areas requiring further exploration will be highlighted to guide future research endeavors and aid in the development of alternative therapeutic strategies to combat surmounting drug resistance in treating MAPK-mediated cancers. VISUAL OVERVIEW: http://mcr.aacrjournals.org/content/molcanres/19/3/361/F1.large.jpg.
Collapse
Affiliation(s)
- Renee Barbosa
- School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lucila A Acevedo
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ronen Marmorstein
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
50
|
Protection of 6-OHDA neurotoxicity by PGF 2α through FP-ERK-Nrf2 signaling in SH-SY5Y cells. Toxicology 2021; 450:152686. [PMID: 33486071 DOI: 10.1016/j.tox.2021.152686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/07/2021] [Accepted: 01/16/2021] [Indexed: 12/26/2022]
Abstract
6-Hydroxydopamine (6-OHDA) is a neurotoxin that destroy dopaminergic neurons and widely used to establish animal models of Parkinson's disease. Prostaglandins (PGs) are involved in various cellular processes, including the damage and repair of neuronal cells. However, the function of PGF2α in neuronal cells remains unclear. In this study, we investigated the effects of PGF2α against 6-OHDA-mediated toxicity in human neuroblastoma SH-SY5Y cells and elucidated its underlying molecular mechanism. When the cells were treated with 6-OHDA (50 μM) for 6 h, the expression levels of PGF2α synthetic enzymes; cyclooxygenase-2 and aldo-keto reductase 1C3 as PGF2α synthase were enhanced in an incubation-time-dependent manner. In addition, the production of PGF2α was increased in 6-OHDA-treated cells. Fluprostenol, a PGF2α receptor (FP) agonist (500 nM), suppressed 6-OHDA-induced cell death by decreasing the production of reactive oxygen species (ROS) and increasing the expression of the anti-oxidant genes. These fluprostenol-mediated effects were inhibited by co-treatment with AL8810, an FP receptor antagonist (1 μM) or transfection with FP siRNA (20 nM). Moreover, 6-OHDA-induced phosphorylation of extracellular signal-regulated kinase (ERK), a member of the mitogen-activated protein kinase family, was inhibited by co-incubation with AL8810. Furthermore, fluprostenol itself enhanced ERK phosphorylation and further elevated the 6-OHDA-induced phosphorylation of ERK. In addition, 6-OHDA induced nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), activating anti-oxidant gene expression, was repressed by co-culturing with AL8810. These results indicate that PGF2α suppressed 6-OHDA-induced neuronal cell death by enhancing anti-oxidant gene expression via the FP receptor-ERK-Nrf2 signaling. Thus, FP receptor is a potential target for inhibition of ROS-mediated neuronal cell death.
Collapse
|