1
|
Cao B, Wu X, Zhou C, Chen H, Xue D, Pan Z. Salvianolic acid A promotes bone-fracture healing via balancing osteoblast and osteoclast differentiation. FASEB J 2025; 39:e70364. [PMID: 39878631 DOI: 10.1096/fj.202402515r] [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: 10/19/2024] [Revised: 01/06/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Nonunion is a significant complication in fracture management for surgeons. Salvianolic acid A (SAA), derived from the traditional Chinese plant Salviae miltiorrhizae Bunge (Danshen), exhibits notable anti-inflammatory and antioxidant properties. Although studies have demonstrated its ability to promote osteogenic differentiation, the exact mechanism of action remains unclear. This study investigated the effects of various SAA concentrations on the osteogenic differentiation of mouse-derived bone marrow mesenchymal stem cells (mBMSCs) and the osteoclastic differentiation of bone marrow-derived macrophages. Our findings indicate that SAA promotes the osteogenic differentiation of mBMSCs in a concentration-dependent manner, primarily by inhibiting the Notch1 signaling pathway. Notably, the administration of two Notch1 agonists (Jagged-1 and VPA) inhibited the effects of SAA on osteogenic differentiation. Additionally, SAA facilitated the autophagic degradation of NICD1, further enhancing osteogenic differentiation. Furthermore, SAA also dose-dependently inhibited the osteoclastic differentiation of bone marrow-derived macrophages, which is linked to its suppression of NF-κB signaling pathways. In a fracture model, SAA demonstrated a capacity to promote healing. In conclusion, SAA enhances bone fracture healing by balancing osteoblast and osteoclast differentiation.
Collapse
Affiliation(s)
- Binhao Cao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, People's Republic of China
| | - Xiaoyong Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, People's Republic of China
| | - Chengwei Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, People's Republic of China
| | - Hongyu Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, People's Republic of China
| | - Deting Xue
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, People's Republic of China
| | - Zhijun Pan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
2
|
Liu D, Liu L, Che X, Wu G. Discovery of paradoxical genes: reevaluating the prognostic impact of overexpressed genes in cancer. Front Cell Dev Biol 2025; 13:1525345. [PMID: 39911323 PMCID: PMC11794808 DOI: 10.3389/fcell.2025.1525345] [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: 11/09/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
Oncogenes are typically overexpressed in tumor tissues and often linked to poor prognosis. However, recent advancements in bioinformatics have revealed that many highly expressed genes in tumors are associated with better patient outcomes. These genes, which act as tumor suppressors, are referred to as "paradoxical genes." Analyzing The Cancer Genome Atlas (TCGA) confirmed the widespread presence of paradoxical genes, and KEGG analysis revealed their role in regulating tumor metabolism. Mechanistically, discrepancies between gene and protein expression-affected by pre- and post-transcriptional modifications-may drive this phenomenon. Mechanisms like upstream open reading frames and alternative splicing contribute to these inconsistencies. Many paradoxical genes modulate the tumor immune microenvironment, exerting tumor-suppressive effects. Further analysis shows that the stage- and tumor-specific expression of these genes, along with their environmental sensitivity, influence their dual roles in various signaling pathways. These findings highlight the importance of paradoxical genes in resisting tumor progression and maintaining cellular homeostasis, offering new avenues for targeted cancer therapy.
Collapse
Affiliation(s)
| | | | - Xiangyu Che
- *Correspondence: Guangzhen Wu, ; Xiangyu Che,
| | | |
Collapse
|
3
|
Tong Y, Wang X, Li R, Xu X, Dai M, Wang N, Fan B, Feng S, Ma T. LSD1 is a promising target to treat cancers by modulating cell stemness. Biochem Pharmacol 2024; 229:116549. [PMID: 39304105 DOI: 10.1016/j.bcp.2024.116549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/15/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
As the first discovered histone demethylase, LSD1 plays a vital role in maintaining pathological processes such as cancer, infection, and immune diseases. Based on previous researches, LSD1 is highly expressed in sorts of tumor cells such as acute myeloid leukemia, non-small cell lung cancer, prostate cancer, breast cancer and gastric cancer, etc. Therefore, targeting LSD1 is a prospective strategy for tumor treatment. Cancer stem cells could preserve self-renewal, cell proliferation, cell migration and malignant phenotype. So, the reduction of tumor cell stemness can effectively inhibit the growth of tumor cells, which may be a new strategy for the treatment of cancers. Up to now, there exist many researches confirming the significant role of LSD1 in regulating the stemness characteristics such as embryonic stem cells differentiation. Many reports show that inhibition of LSD1 effectively decreases the property of cancer cell stemness. However, there lacks a detailed review about the relationship between LSD1 and cancer cell stemness. Herein, in this review, we summarized the mechanisms how LSD1 regulates cell stemness comprehensively. In addition, some related inhibitors targeting LSD1 to reduce the proliferation characteristics of cancer stem cells are also described.
Collapse
Affiliation(s)
- Yaoyuan Tong
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Xiaoru Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ruonan Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Xiangyu Xu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Mengge Dai
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Nan Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Boyi Fan
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Siqi Feng
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Ting Ma
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
4
|
Dakal TC, Bhushan R, Xu C, Gadi BR, Cameotra SS, Yadav V, Maciaczyk J, Schmidt‐Wolf IGH, Kumar A, Sharma A. Intricate relationship between cancer stemness, metastasis, and drug resistance. MedComm (Beijing) 2024; 5:e710. [PMID: 39309691 PMCID: PMC11416093 DOI: 10.1002/mco2.710] [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: 03/13/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/25/2024] Open
Abstract
Cancer stem cells (CSCs) are widely acknowledged as the drivers of tumor initiation, epithelial-mesenchymal transition (EMT) progression, and metastasis. Originating from both hematologic and solid malignancies, CSCs exhibit quiescence, pluripotency, and self-renewal akin to normal stem cells, thus orchestrating tumor heterogeneity and growth. Through a dynamic interplay with the tumor microenvironment (TME) and intricate signaling cascades, CSCs undergo transitions from differentiated cancer cells, culminating in therapy resistance and disease recurrence. This review undertakes an in-depth analysis of the multifaceted mechanisms underlying cancer stemness and CSC-mediated resistance to therapy. Intrinsic factors encompassing the TME, hypoxic conditions, and oxidative stress, alongside extrinsic processes such as drug efflux mechanisms, collectively contribute to therapeutic resistance. An exploration into key signaling pathways, including JAK/STAT, WNT, NOTCH, and HEDGEHOG, sheds light on their pivotal roles in sustaining CSCs phenotypes. Insights gleaned from preclinical and clinical studies hold promise in refining drug discovery efforts and optimizing therapeutic interventions, especially chimeric antigen receptor (CAR)-T cell therapy, cytokine-induced killer (CIK) cell therapy, natural killer (NK) cell-mediated CSC-targeting and others. Ultimately use of cell sorting and single cell sequencing approaches for elucidating the fundamental characteristics and resistance mechanisms inherent in CSCs will enhance our comprehension of CSC and intratumor heterogeneity, which ultimately would inform about tailored and personalized interventions.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology LabDepartment of BiotechnologyMohanlal Sukhadia UniversityUdaipurRajasthanIndia
| | - Ravi Bhushan
- Department of ZoologyM.S. CollegeMotihariBiharIndia
| | - Caiming Xu
- Department of General SurgeryThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research InstituteCity of HopeMonroviaCaliforniaUSA
| | - Bhana Ram Gadi
- Stress Physiology and Molecular Biology LaboratoryDepartment of BotanyJai Narain Vyas UniversityJodhpurRajasthanIndia
| | | | - Vikas Yadav
- School of Life SciencesJawaharlal Nehru UniversityNew DelhiIndia
| | - Jarek Maciaczyk
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
| | - Ingo G. H. Schmidt‐Wolf
- Center for Integrated Oncology (CIO)Department of Integrated OncologyUniversity Hospital BonnBonnGermany
| | - Abhishek Kumar
- Manipal Academy of Higher EducationManipalKarnatakaIndia
- Institute of BioinformaticsInternational Technology ParkBangaloreIndia
| | - Amit Sharma
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
- Center for Integrated Oncology (CIO)Department of Integrated OncologyUniversity Hospital BonnBonnGermany
| |
Collapse
|
5
|
Shimojo H, Masaki T, Kageyama R. The Neurog2-Tbr2 axis forms a continuous transition to the neurogenic gene expression state in neural stem cells. Dev Cell 2024; 59:1913-1923.e6. [PMID: 38772376 DOI: 10.1016/j.devcel.2024.04.019] [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: 02/04/2023] [Revised: 03/04/2024] [Accepted: 04/28/2024] [Indexed: 05/23/2024]
Abstract
Neural stem cells (NSCs) differentiate into neuron-fated intermediate progenitor cells (IPCs) via cell division. Although differentiation from NSCs to IPCs is a discrete process, recent transcriptome analyses identified a continuous transcriptional trajectory during this process, raising the question of how to reconcile these contradictory observations. In mouse NSCs, Hes1 expression oscillates, regulating the oscillatory expression of the proneural gene Neurog2, while Hes1 expression disappears in IPCs. Thus, the transition from Hes1 oscillation to suppression is involved in the differentiation of NSCs to IPCs. Here, we found that Neurog2 oscillations induce the accumulation of Tbr2, which suppresses Hes1 expression, generating an IPC-like gene expression state in NSCs. In the absence of Tbr2, Hes1 expression is up-regulated, decreasing the formation of IPCs. These results indicate that the Neurog2-Tbr2 axis forms a continuous transcriptional trajectory to an IPC-like neurogenic state in NSCs, which then differentiate into IPCs via cell division.
Collapse
Affiliation(s)
- Hiromi Shimojo
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan.
| | - Taimu Masaki
- RIKEN Center for Brain Science, Wako 351-0198, Japan; Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Ryoichiro Kageyama
- RIKEN Center for Brain Science, Wako 351-0198, Japan; Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
6
|
Sachan N, Sharma V, Mutsuddi M, Mukherjee A. Notch signalling: multifaceted role in development and disease. FEBS J 2024; 291:3030-3059. [PMID: 37166442 DOI: 10.1111/febs.16815] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/08/2023] [Accepted: 05/10/2023] [Indexed: 05/12/2023]
Abstract
Notch pathway is an evolutionarily conserved signalling system that operates to influence an astonishing array of cell fate decisions in different developmental contexts. Notch signalling plays important roles in many developmental processes, making it difficult to name a tissue or a developing organ that does not depend on Notch function at one stage or another. Thus, dysregulation of Notch signalling is associated with many developmental defects and various pathological conditions, including cancer. Although many recent advances have been made to reveal different aspects of the Notch signalling mechanism and its intricate regulation, there are still many unanswered questions related to how the Notch signalling pathway functions in so many developmental events. The same pathway can be deployed in numerous cellular contexts to play varied and critical roles in an organism's development and this is only possible because of the complex regulatory mechanisms of the pathway. In this review, we provide an overview of the mechanism and regulation of the Notch signalling pathway along with its multifaceted functions in different aspects of development and disease.
Collapse
Affiliation(s)
- Nalani Sachan
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
7
|
Li L, Yang W, Pan Y, Ye R, Wang Y, Li S, Jiang H, Zhang Q, Wang X, Yan J. Chidamide enhances T-cell-mediated anti-tumor immune function by inhibiting NOTCH1/NFATC1 signaling pathway in ABC-type diffuse large B-cell lymphoma. Leuk Lymphoma 2024; 65:895-910. [PMID: 38497543 DOI: 10.1080/10428194.2024.2328227] [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: 12/11/2023] [Accepted: 03/03/2024] [Indexed: 03/19/2024]
Abstract
Chidamide (CS055/HBI-8000, tucidinostat) has shown promising effects in the clinical treatment of various hematologic tumors. Diffuse large B-cell lymphoma (DLBCL) has shown highly heterogeneous biological characteristics. There are complex mechanisms of the role of chidamide in DLBCL for in-depth study. It is essential to probe further into the mechanism of drug-tumor interactions as a guide to clinical application and to understand the occurrence and progression of DLBCL. In vitro and in vivo models were utilized to determine the effects of chidamide on signaling pathways involved in the DLBCL tumor microenvironment. The experimental results show that chidamide inhibited the proliferation of DLBCL cell lines in a dose- and time-dependent manner, and down-regulated the expression of NOTCH1 and NFATC1 in DLBCL cells as well as decreased the concentration of IL-10 in the supernatant. In addition, chidamide significantly lowered the expression of PD1 or TIM3 on CD4+T cells and CD8+T cells and elevated the levels of IL-2, IFN-γ, and TNF-α in the serum of animal models, which augmented the function of circulating T cells and tumor-infiltrating T cells and ultimately significantly repressed the growth of tumors. These findings prove that chidamide can effectively inhibit the cell activity of DLBCL cell lines by inhibiting the activation of NOTCH1 and NFATC1 signaling pathways. It can also improve the abnormal DLBCL microenvironment in which immune escape occurs, and inhibit immune escape. This study provides a new therapeutic idea for the exploration of individualized precision therapy for patients with malignant lymphoma.
Collapse
MESH Headings
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Humans
- NFATC Transcription Factors/metabolism
- Receptor, Notch1/metabolism
- Receptor, Notch1/genetics
- Aminopyridines/pharmacology
- Aminopyridines/therapeutic use
- Signal Transduction/drug effects
- Benzamides/pharmacology
- Benzamides/therapeutic use
- Animals
- Mice
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Cell Proliferation/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/drug effects
- T-Lymphocytes/metabolism
- Disease Models, Animal
Collapse
Affiliation(s)
- Li Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Wenjing Yang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Yuanyuan Pan
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Ruyu Ye
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Yu Wang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Sijia Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Haoyan Jiang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Qi Zhang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Xiaobo Wang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Jinsong Yan
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| |
Collapse
|
8
|
Meng X, Bai X, Ke A, Li K, Lei Y, Ding S, Dai D. Long Non-Coding RNAs in Drug Resistance of Gastric Cancer: Complex Mechanisms and Potential Clinical Applications. Biomolecules 2024; 14:608. [PMID: 38927012 PMCID: PMC11201466 DOI: 10.3390/biom14060608] [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: 03/10/2024] [Revised: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Gastric cancer (GC) ranks as the third most prevalent malignancy and a leading cause of cancer-related mortality worldwide. However, the majority of patients with GC are diagnosed at an advanced stage, highlighting the urgent need for effective perioperative and postoperative chemotherapy to prevent relapse and metastasis. The current treatment strategies have limited overall efficacy because of intrinsic or acquired drug resistance. Recent evidence suggests that dysregulated long non-coding RNAs (lncRNAs) play a significant role in mediating drug resistance in GC. Therefore, there is an imperative to explore novel molecular mechanisms underlying drug resistance in order to overcome this challenging issue. With advancements in deep transcriptome sequencing technology, lncRNAs-once considered transcriptional noise-have garnered widespread attention as potential regulators of carcinogenesis, including tumor cell proliferation, metastasis, and sensitivity to chemo- or radiotherapy through multiple regulatory mechanisms. In light of these findings, we aim to review the mechanisms by which lncRNAs contribute to drug therapy resistance in GC with the goal of providing new insights and breakthroughs toward overcoming this formidable obstacle.
Collapse
Affiliation(s)
- Xiangyu Meng
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China
| | - Xiao Bai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
| | - Angting Ke
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
| | - Kaiqiang Li
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
| | - Yun Lei
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
| | - Siqi Ding
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
| | - Dongqiu Dai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; (X.M.); (X.B.); (K.L.); (Y.L.); (S.D.)
- Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| |
Collapse
|
9
|
Kacker S, Parsad V, Singh N, Hordiichuk D, Alvarez S, Gohar M, Kacker A, Rai SK. Planar Cell Polarity Signaling: Coordinated Crosstalk for Cell Orientation. J Dev Biol 2024; 12:12. [PMID: 38804432 PMCID: PMC11130840 DOI: 10.3390/jdb12020012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/08/2024] [Accepted: 04/13/2024] [Indexed: 05/29/2024] Open
Abstract
The planar cell polarity (PCP) system is essential for positioning cells in 3D networks to establish the proper morphogenesis, structure, and function of organs during embryonic development. The PCP system uses inter- and intracellular feedback interactions between components of the core PCP, characterized by coordinated planar polarization and asymmetric distribution of cell populations inside the cells. PCP signaling connects the anterior-posterior to left-right embryonic plane polarity through the polarization of cilia in the Kupffer's vesicle/node in vertebrates. Experimental investigations on various genetic ablation-based models demonstrated the functions of PCP in planar polarization and associated genetic disorders. This review paper aims to provide a comprehensive overview of PCP signaling history, core components of the PCP signaling pathway, molecular mechanisms underlying PCP signaling, interactions with other signaling pathways, and the role of PCP in organ and embryonic development. Moreover, we will delve into the negative feedback regulation of PCP to maintain polarity, human genetic disorders associated with PCP defects, as well as challenges associated with PCP.
Collapse
Affiliation(s)
- Sandeep Kacker
- Department of Pharmacology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis;
| | - Varuneshwar Parsad
- Department of Human Body Structure and Function, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (V.P.); (D.H.)
| | - Naveen Singh
- Department of Cerll and Molecular Biology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (N.S.); (S.A.); (M.G.)
| | - Daria Hordiichuk
- Department of Human Body Structure and Function, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (V.P.); (D.H.)
| | - Stacy Alvarez
- Department of Cerll and Molecular Biology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (N.S.); (S.A.); (M.G.)
| | - Mahnoor Gohar
- Department of Cerll and Molecular Biology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (N.S.); (S.A.); (M.G.)
| | - Anshu Kacker
- Department of Histology and Human Physiology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis;
| | - Sunil Kumar Rai
- Department of Cerll and Molecular Biology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (N.S.); (S.A.); (M.G.)
| |
Collapse
|
10
|
Verma D, Singh A, Singh J, Mutsuddi M, Mukherjee A. Regulation of Notch signaling by non-muscle myosin II Zipper in Drosophila. Cell Mol Life Sci 2024; 81:195. [PMID: 38653877 PMCID: PMC11039529 DOI: 10.1007/s00018-024-05142-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/27/2023] [Accepted: 01/23/2024] [Indexed: 04/25/2024]
Abstract
The Notch pathway is an evolutionarily conserved signaling system that is intricately regulated at multiple levels and it influences different aspects of development. In an effort to identify novel components involved in Notch signaling and its regulation, we carried out protein interaction screens which identified non-muscle myosin II Zipper (Zip) as an interacting partner of Notch. Physical interaction between Notch and Zip was further validated by co-immunoprecipitation studies. Immunocytochemical analyses revealed that Notch and Zip co-localize within same cytoplasmic compartment. Different alleles of zip also showed strong genetic interactions with Notch pathway components. Downregulation of Zip resulted in wing phenotypes that were reminiscent of Notch loss-of-function phenotypes and a perturbed expression of Notch downstream targets, Cut and Deadpan. Further, synergistic interaction between Notch and Zip resulted in highly ectopic expression of these Notch targets. Activated Notch-induced tumorous phenotype of larval tissues was enhanced by over-expression of Zip. Notch-Zip synergy resulted in the activation of JNK pathway that consequently lead to MMP activation and proliferation. Taken together, our results suggest that Zip may play an important role in regulation of Notch signaling.
Collapse
Affiliation(s)
- Dipti Verma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Ankita Singh
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Jyoti Singh
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
11
|
Sudhadevi T, Ackerman SJ, Jafri A, Basa P, Ha AW, Natarajan V, Harijith A. Sphingosine kinase 1-specific inhibitor PF543 reduces goblet cell metaplasia of bronchial epithelium in an acute asthma model. Am J Physiol Lung Cell Mol Physiol 2024; 326:L377-L392. [PMID: 38290992 PMCID: PMC11281799 DOI: 10.1152/ajplung.00269.2023] [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: 08/22/2023] [Revised: 01/16/2024] [Accepted: 01/21/2024] [Indexed: 02/01/2024] Open
Abstract
Sphingosine kinase 1 (SPHK1) has been shown to play a key role in the pathogenesis of asthma where SPHK1-generated sphingosine-1-phosphate (S1P) is known to mediate innate and adaptive immunity while promoting mast cell degranulation. Goblet cell metaplasia (GCM) contributes to airway obstruction in asthma and has been demonstrated in animal models. We investigated the role of PF543, a SPHK1-specific inhibitor, in preventing the pathogenesis of GCM using a murine (C57BL/6) model of allergen-induced acute asthma. Treatment with PF543 before triple allergen exposure (DRA: House dust mite, Ragweed pollen, and Aspergillus) reduced inflammation, eosinophilic response, and GCM followed by reduced airway hyperreactivity to intravenous methacholine. Furthermore, DRA exposure was associated with increased expression of SPHK1 in the airway epithelium which was reduced by PF543. DRA-induced reduction of acetylated α-tubulin in airway epithelium was associated with an increased expression of NOTCH2 and SPDEF which was prevented by PF543. In vitro studies using human primary airway epithelial cells showed that inhibition of SPHK1 using PF543 prevented an allergen-induced increase of both NOTCH2 and SPDEF. siRNA silencing of SPHK1 prevented the allergen-induced increase of both NOTCH2 and SPDEF. NOTCH2 silencing was associated with a reduction of SPDEF but not that of SPHK1 upon allergen exposure. Our studies demonstrate that inhibition of SPHK1 protected allergen-challenged airways by preventing GCM and airway hyperreactivity, associated with downregulation of the NOTCH2-SPDEF signaling pathway. This suggests a potential novel link between SPHK1, GCM, and airway remodeling in asthma.NEW & NOTEWORTHY The role of SPHK1-specific inhibitor, PF543, in preventing goblet cell metaplasia (GCM) and airway hyperreactivity (AHR) is established in an allergen-induced mouse model. This protection was associated with the downregulation of NOTCH2-SPDEF signaling pathway, suggesting a novel link between SPHK1, GCM, and AHR.
Collapse
Affiliation(s)
- Tara Sudhadevi
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States
| | - Steven J Ackerman
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Anjum Jafri
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States
| | - Prathima Basa
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States
| | - Alison W Ha
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Viswanathan Natarajan
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Anantha Harijith
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
12
|
Zafar S, Fatima SI, Schmitz M, Zerr I. Current Technologies Unraveling the Significance of Post-Translational Modifications (PTMs) as Crucial Players in Neurodegeneration. Biomolecules 2024; 14:118. [PMID: 38254718 PMCID: PMC10813409 DOI: 10.3390/biom14010118] [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: 12/14/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Neurodegenerative disorders, such as Parkinson's disease, Alzheimer's disease, and Huntington's disease, are identified and characterized by the progressive loss of neurons and neuronal dysfunction, resulting in cognitive and motor impairment. Recent research has shown the importance of PTMs, such as phosphorylation, acetylation, methylation, ubiquitination, sumoylation, nitration, truncation, O-GlcNAcylation, and hydroxylation, in the progression of neurodegenerative disorders. PTMs can alter protein structure and function, affecting protein stability, localization, interactions, and enzymatic activity. Aberrant PTMs can lead to protein misfolding and aggregation, impaired degradation, and clearance, and ultimately, to neuronal dysfunction and death. The main objective of this review is to provide an overview of the PTMs involved in neurodegeneration, their underlying mechanisms, methods to isolate PTMs, and the potential therapeutic targets for these disorders. The PTMs discussed in this article include tau phosphorylation, α-synuclein and Huntingtin ubiquitination, histone acetylation and methylation, and RNA modifications. Understanding the role of PTMs in neurodegenerative diseases may provide new therapeutic strategies for these devastating disorders.
Collapse
Affiliation(s)
- Saima Zafar
- Department of Neurology, Clinical Dementia Center and DZNE, University Medical Center Goettingen (UMG), Georg-August University, Robert-Koch-Str. 40, 37075 Goettingen, Germany
- Biomedical Engineering and Sciences Department, School of Mechanical and Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Bolan Road, H-12, Islamabad 44000, Pakistan
| | - Shehzadi Irum Fatima
- Department of Neurology, Clinical Dementia Center and DZNE, University Medical Center Goettingen (UMG), Georg-August University, Robert-Koch-Str. 40, 37075 Goettingen, Germany
| | - Matthias Schmitz
- Department of Neurology, Clinical Dementia Center and DZNE, University Medical Center Goettingen (UMG), Georg-August University, Robert-Koch-Str. 40, 37075 Goettingen, Germany
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center and DZNE, University Medical Center Goettingen (UMG), Georg-August University, Robert-Koch-Str. 40, 37075 Goettingen, Germany
| |
Collapse
|
13
|
Cai J, Qiao Y, Chen L, Lu Y, Zheng D. Regulation of the Notch signaling pathway by natural products for cancer therapy. J Nutr Biochem 2024; 123:109483. [PMID: 37848105 DOI: 10.1016/j.jnutbio.2023.109483] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/13/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023]
Abstract
The Notch signaling pathway is an evolutionarily conserved pathway that modulates normal biological processes involved in cellular differentiation, apoptosis, and stem cell self-renewal in a context-dependent fashion. Attributed to its pleiotropic physiological roles, both overexpression and silencing of the pathway are associated with the emergence, progression, and poorer prognosis in various types of cancer. To decrease disease incidence and promote survival, targeting Notch may have chemopreventive and anti-cancer effects. Natural products with profound historical origins have distinguished themselves from other therapies due to their easy access, high biological compatibility, low toxicity, and reliable effects at specific physiological sites in vivo. This review describes the Notch signaling pathway, particularly its normal activation process, and some main illnesses related to Notch signaling pathway dysregulation. Emphasis is placed on the effects and mechanisms of natural products targeting the Notch signaling pathway in diverse cancer types, including curcumin, ellagic acid (EA), resveratrol, genistein, epigallocatechin-3-gallate (EGCG), quercetin, and xanthohumol and so on. Existing evidence indicates that natural products are feasible solution to fight against cancer by targeting Notch signaling, either alone or in combination with current therapeutic agents.
Collapse
Affiliation(s)
- Jiayi Cai
- School of Stomatology, Fujian Medical University, Fuzhou 350122, China
| | - Yajie Qiao
- School of Stomatology, Fujian Medical University, Fuzhou 350122, China
| | - Lingbin Chen
- School of Stomatology, Fujian Medical University, Fuzhou 350122, China
| | - Youguang Lu
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China; Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350001, China
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China.
| |
Collapse
|
14
|
Shekar N, Vuong P, Kaur P. Analysing potent biomarkers along phytochemicals for breast cancer therapy: an in silico approach. Breast Cancer Res Treat 2024; 203:29-47. [PMID: 37726449 PMCID: PMC10771382 DOI: 10.1007/s10549-023-07107-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE This research focused on the identification of herbal compounds as potential anti-cancer drugs, especially for breast cancer, that involved the recognition of Notch downstream targets NOTCH proteins (1-4) specifically expressed in breast tumours as biomarkers for prognosis, along with P53 tumour antigens, that were used as comparisons to check the sensitivity of the herbal bio-compounds. METHODS After investigating phytochemical candidates, we employed an approach for computer-aided drug design and analysis to find strong breast cancer inhibitors. The present study utilized in silico analyses and protein docking techniques to characterize and rank selected bio-compounds for their efficiency in oncogenic inhibition for use in precise carcinomic cell growth control. RESULTS Several of the identified phytocompounds found in herbs followed Lipinski's Rule of Five and could be further investigated as potential medicinal molecules. Based on the Vina score obtained after the docking process, the active compound Epigallocatechin gallate in green tea with NOTCH (1-4) and P53 proteins showed promising results for future drug repurposing. The stiffness and binding stability of green tea pharmacological complexes were further elucidated by the molecular dynamic simulations carried out for the highest scoring phytochemical ligand complex. CONCLUSION The target-ligand complex of green tea active compound Epigallocatechin gallate with NOTCH (1-4) had the potential to become potent anti-breast cancer therapeutic candidates following further research involving wet-lab experiments.
Collapse
Affiliation(s)
- Nivruthi Shekar
- UWA School of Agriculture and Environment, University of Western Australia, 35-Stirling Highway, Perth, WA, 6009, Australia
| | - Paton Vuong
- UWA School of Agriculture and Environment, University of Western Australia, 35-Stirling Highway, Perth, WA, 6009, Australia
| | - Parwinder Kaur
- UWA School of Agriculture and Environment, University of Western Australia, 35-Stirling Highway, Perth, WA, 6009, Australia.
| |
Collapse
|
15
|
Corcoran E, Olayinka A, di Luca M, Gusti Y, Hakimjavadi R, O'Connor B, Redmond EM, Cahill PA. N-Glycans on the extracellular domain of the Notch1 receptor control Jagged-1 induced Notch signalling and myogenic differentiation of S100β resident vascular stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567576. [PMID: 38014317 PMCID: PMC10680845 DOI: 10.1101/2023.11.17.567576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Notch signalling, critical for development and postnatal homeostasis of the vascular system, is highly regulated by several mechanisms including glycosylation. While the importance of O-linked glycosylation is widely accepted, the structure and function of N-glycans has yet to be defined. Here, we take advantage of lectin binding assays in combination with pharmacological, molecular, and site-directed mutagenetic approaches to study N-glycosylation of the Notch1 receptor. We find that several key oligosaccharides containing bisecting or core fucosylated structures decorate the receptor, control expression and receptor trafficking, and dictate Jagged-1 activation of Notch target genes and myogenic differentiation of multipotent S100β vascular stem cells. N-glycans at asparagine (N) 1241 and 1587 protect the receptor from accelerated degradation, while the oligosaccharide at N888 directly affects signal transduction. Conversely, N-linked glycans at N959, N1179, N1489 do not impact canonical signalling but inhibit differentiation. Our work highlights a novel functional role for N-glycans in controlling Notch1 signalling and differentiation of vascular stem cells.
Collapse
Affiliation(s)
- Eoin Corcoran
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Abidemi Olayinka
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Mariana di Luca
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Yusof Gusti
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Roya Hakimjavadi
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Brendan O'Connor
- School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Eileen M Redmond
- Department of Surgery, University of Rochester, 601 Elmwood Ave, Rochester, NY 14642, United States
| | - Paul A Cahill
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| |
Collapse
|
16
|
Dietrich B, Kunihs V, Lackner AI, Meinhardt G, Koo BK, Pollheimer J, Haider S, Knöfler M. NOTCH3 signalling controls human trophoblast stem cell expansion and differentiation. Development 2023; 150:dev202152. [PMID: 37905445 DOI: 10.1242/dev.202152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
Failures in growth and differentiation of the early human placenta are associated with severe pregnancy disorders such as pre-eclampsia and fetal growth restriction. However, regulatory mechanisms controlling development of placental epithelial cells, the trophoblasts, remain poorly elucidated. Using trophoblast stem cells (TSCs), trophoblast organoids (TB-ORGs) and primary cytotrophoblasts (CTBs) of early pregnancy, we herein show that autocrine NOTCH3 signalling controls human placental expansion and differentiation. The NOTCH3 receptor was specifically expressed in proliferative CTB progenitors and its active form, the nuclear NOTCH3 intracellular domain (NOTCH3-ICD), interacted with the transcriptional co-activator mastermind-like 1 (MAML1). Doxycycline-inducible expression of dominant-negative MAML1 in TSC lines provoked cell fusion and upregulation of genes specific for multinucleated syncytiotrophoblasts, which are the differentiated hormone-producing cells of the placenta. However, progenitor expansion and markers of trophoblast stemness and proliferation were suppressed. Accordingly, inhibition of NOTCH3 signalling diminished growth of TB-ORGs, whereas overexpression of NOTCH3-ICD in primary CTBs and TSCs showed opposite effects. In conclusion, the data suggest that canonical NOTCH3 signalling plays a key role in human placental development by promoting self-renewal of CTB progenitors.
Collapse
Affiliation(s)
- Bianca Dietrich
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Victoria Kunihs
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Andreas I Lackner
- Maternal-Fetal Immunology Group, Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, A-1090 Vienna, Austria
| | - Gudrun Meinhardt
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Science, Yuseong-Gu, Daejeon 34126, Republic of Korea
| | - Jürgen Pollheimer
- Maternal-Fetal Immunology Group, Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sandra Haider
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Martin Knöfler
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
17
|
Ma Z, Zeng Y, Wang M, Liu W, Zhou J, Wu C, Hou L, Yin B, Qiang B, Shu P, Peng X. N4BP1 mediates RAM domain-dependent notch signaling turnover during neocortical development. EMBO J 2023; 42:e113383. [PMID: 37807845 PMCID: PMC10646556 DOI: 10.15252/embj.2022113383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 10/10/2023] Open
Abstract
Notch signaling pathway activity, particularly fluctuations in the biologically active effector fragment NICD, is required for rapid and efficient dynamic regulation of proper fate decisions in stem cells. In this study, we identified NEDD4-binding protein 1 (N4BP1), which is highly expressed in the developing mouse cerebral cortex, as a negative modulator of Notch signaling dynamics in neural progenitor cells. Intriguingly, N4BP1 regulated NICD stability specifically after Notch1 S3 cleavage through ubiquitin-mediated degradation that depended on its RAM domain, not its PEST domain, as had been extensively and previously described. The CoCUN domain in N4BP1, particularly the "Phe-Pro" motif (862/863 amino acid), was indispensable for mediating NICD degradation. The Ring family E3 ligase Trim21 was, in contrast to other NEDD4 family members, required for N4BP1-regulated NICD degradation. Overexpression of N4BP1 in cortical neural progenitors promoted neural stem cell differentiation, whereas neural progenitor cells lacking N4BP1 were sensitized to Notch signaling, resulting in the maintenance of stem-like properties in neural progenitor cells and lower production of cortical neurons.
Collapse
Affiliation(s)
- Zhihua Ma
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Yi Zeng
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Present address:
Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)The Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Ming Wang
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Present address:
Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren HospitalCapital Medical University, Beijing Key Laboratory of Nasal Diseases, Beijing Institute of OtolaryngologyBeijingChina
| | - Wei Liu
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Jiafeng Zhou
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Chao Wu
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Lin Hou
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
| | - Bin Yin
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
| | - Boqin Qiang
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
| | - Pengcheng Shu
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
- Chinese Institute for Brain ResearchBeijingChina
| | - Xiaozhong Peng
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Respiratory Health and MultimorbidityBeijingChina
- Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| |
Collapse
|
18
|
Dong S, Wang Z, Xiong W. POFUT1 promotes gastric cancer progression through Notch/Wnt dual signaling pathways dependent on the parafibromin-NICD1-β-catenin complex. J Chin Med Assoc 2023; 86:806-817. [PMID: 37501238 DOI: 10.1097/jcma.0000000000000957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Aberrant glycosylation performed by glycosyltransferases is a leading cause of gastric cancer (GC). Protein O-fucosyltransferase 1 (POFUT1) expression is increased in GC specimens and cells. In this study, the biological effects and mechanisms of POFUT1 underlying the development of GC were investigated. METHODS POFUT1 downregulated and upregulated GC cells were established. The effects of POFUT1 on cell proliferation, metastasis and apoptosis were examined using cell counting kit-8 (CCK8) assay, transwell assay, and flow cytometry. Subcutaneous xenograft tumor models were established followed by immunohistochemistry staining of resected tumors. Facilitating modulators and transcription factors were detected by western blot, immunofluorescence, luciferase reporter assay, and co-immunoprecipitation. RESULTS POFUT1 played a pro-oncogenic role both in vivo and in vitro, which promoted proliferation and metastasis, as well as inhibited apoptosis in GC cells. POFUT1 promoted Cyclin D3 expression and inhibited the expression of apoptotic proteins, such as Bcl-2-associated X protein (Bax) and cleaved caspase 3, facilitating tumor growth. Moreover, POFUT1 accelerated matrix metalloproteases expression and attenuated E-cadherin expression, contributing to GC metastasis. In addition, POFUT1 expression promoted the expression and nuclear translocation of Notch1 intracellular domain (NICD1) and β-catenin and inhibited β-catenin phosphorylation degradation, accompanied by the activation of recombination signal binding protein-Jκ (RBP-J) and T-cell factor (TCF) transcription factors, respectively. It is notable that parafibromin integrated NICD1 and β-catenin, enabling the concerted activation of Wnt and Notch signaling targeted proteins. CONCLUSION These observations indicated that POFUT1 promoted GC development through activation of Notch and Wnt signaling pathways, which depended on the parafibromin-NICD1-β-catenin complex. This work provides new evidence for the further diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Shuang Dong
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhirong Wang
- Department of Gastroenterology, Shanghai Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Wujun Xiong
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
19
|
Mukherjee M, Levine H. The alternate ligand Jagged enhances the robustness of Notch signaling patterns. SOFT MATTER 2023. [PMID: 37323020 DOI: 10.1039/d2sm01508k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The Notch pathway, an example of juxtacrine signaling, is an evolutionary conserved cell-cell communication mechanism. It governs emergent spatiotemporal patterning in tissues during development, wound healing and tumorigenesis. Communication occurs when Notch receptors of one cell bind to either of its ligands, Delta/Jagged of the neighboring cell. In general, Delta-mediated signaling drives neighboring cells to have an opposite fate (lateral inhibition) whereas Jagged-mediated signaling drives cells to maintain similar fates (lateral induction). Here, by deriving and solving a reduced set of 12 coupled ordinary differential equations for the Notch-Delta-Jagged system on a hexagonal grid of cells, we determine the allowed states across different parameter sets. We also show that Jagged (at low dose) acts synergistically with Delta to enable more robust pattern formation by making the neighboring cell states more distinct from each other, despite its lateral induction property. Our findings extend our understanding of the possible synergistic role of Jagged with Delta which had been previously proposed through experiments and models in the context of chick inner ear development. Finally, we show how Jagged can help to expand the bistable (both uniform and hexagon phases are stable) region, where a local perturbation can spread over time in an ordered manner to create a biologically relevant, perfectly ordered lateral inhibition pattern.
Collapse
Affiliation(s)
- Mrinmoy Mukherjee
- Center for Theoretical Biological Physics, Northeastern University, Boston, MA, USA.
| | - Herbert Levine
- Center for Theoretical Biological Physics, Northeastern University, Boston, MA, USA.
- Depts. of Physics and Bioengineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
20
|
Lu C, Zhang J, Wang B, Gao Q, Ma K, Pei S, Li J, Cui S. Casein kinase 1α is required to maintain murine hypothalamic pro-opiomelanocortin expression. iScience 2023; 26:106670. [PMID: 37168577 PMCID: PMC10165255 DOI: 10.1016/j.isci.2023.106670] [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: 12/13/2022] [Revised: 02/08/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Hypothalamic pro-opiomelanocortin (POMC) neuron development is considered to play an essential role in the development of obesity. However, the underlying mechanisms remain unclear. Casein kinase 1α (CK1α) was expressed in the embryonic mouse hypothalamus at high levels and colocalized with POMC neurons. CK1α deletion in POMC neurons caused weight gain, metabolic defects, and increased food intake. The number of POMC-expressing cells was considerably decreased in Csnk1a1fl/fl;POMCcre (PKO) mice from embryonic day 15.5 to postnatal day 60, while apoptosis of POMC neurons was not affected. Furthermore, unchanged POMC progenitor cells and a decreased POMC phenotype established CK1α function in hypothalamic POMC neuron development. CK1α deletion led to elevated Notch intracellular domain (NICD) protein expression, and NICD inhibition rescued the PKO mouse phenotype. In summary, CK1α is involved in hypothalamic POMC expression via NICD-POMC signaling, deepening our understanding of POMC neuron development and control of systemic metabolic functions.
Collapse
Affiliation(s)
- Chenyang Lu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Jinglin Zhang
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Bingjie Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Qiao Gao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Kezhe Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Shaona Pei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Juxue Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People’s Republic of China
- Corresponding author
| |
Collapse
|
21
|
Bai MR, Pei HY, Zhou Y, Song HL, Pan WH, Gong YM, Wu WJ, Yu WW, Cui MM, Gu BL, Chu X, Cai W. Association analysis and functional follow-up identified common variants of JAG1 accounting for risk to biliary atresia. Front Genet 2023; 14:1186882. [PMID: 37255715 PMCID: PMC10225652 DOI: 10.3389/fgene.2023.1186882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/05/2023] [Indexed: 06/01/2023] Open
Abstract
Background: Biliary atresia (BA) is a destructive, obliterative cholangiopathy characterized by progressive fibro-inflammatory disorder and obliteration of intra- and extrahepatic bile ducts. The Jagged1 (JAG1) gene mutations have been found in some isolated BA cases. We aim to explore the association of common variants in JAG1 with isolated BA risk in the Chinese Han population. Methods: We genotyped 31 tag single nucleotide polymorphisms covering the JAG1 gene region in 333 BA patients and 1,665 healthy controls from the Chinese population, and performed case-control association analysis. The expression patterns of JAG1 homologs were investigated in zebrafish embryos, and the roles of jag1a and jag1b in biliary development were examined by morpholino knockdown in zebrafish. Results: Single nucleotide polymorphisms rs6077861 [P Allelic = 1.74 × 10-4, odds ratio = 1.78, 95% confidence interval: 1.31-2.40] and rs3748478 (P Allelic = 5.77 × 10-4, odds ratio = 1.39, 95% confidence interval: 1.15-1.67) located in the intron region of JAG1 showed significant associations with BA susceptibility. The JAG1 homologs, jag1a and jag1b genes were expressed in the developing hepatobiliary duct of zebrafish, especially at 72 and 96 h postfertilization. Knockdown of both jag1a and jag1b led to poor biliary secretion, sparse intrahepatic bile duct network and smaller or no gallbladders compared with control embryos in the zebrafish model. Conclusion: Common genetic variants of JAG1 were associated with BA susceptibility. Knockdown of JAG1 homologs led to defective intrahepatic and extrahepatic bile ducts in zebrafish. These results suggest that JAG1 might be implicated in the etiology of BA.
Collapse
Affiliation(s)
- Mei-Rong Bai
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Hao-Yue Pei
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Ying Zhou
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huan-Lei Song
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Wei-Hua Pan
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi-Ming Gong
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wen-Jie Wu
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wen-Wen Yu
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Meng-Meng Cui
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Bei-Lin Gu
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Xun Chu
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Wei Cai
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Institute of Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| |
Collapse
|
22
|
Hull KL, Greenwood MP, Lloyd M, Bester-van der Merwe AE, Rhode C. Gene expression differentials driven by mass rearing and artificial selection in black soldier fly colonies. INSECT MOLECULAR BIOLOGY 2023; 32:86-105. [PMID: 36322045 DOI: 10.1111/imb.12816] [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: 02/03/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
The micro-evolutionary forces that shape genetic diversity during domestication have been assessed in many plant and animal systems. However, the impact of these processes on gene expression, and consequent functional adaptation to artificial environments, remains under-investigated. In this study, whole-transcriptome dynamics associated with the early stages of domestication of the black soldier fly (BSF), Hermetia illucens, were assessed. Differential gene expression (DGE) was evaluated in relation to (i) generational time within the cultured environment (F2 vs. F3), and (ii) two selection strategies [no artificial selective pressure (NS); and selection for greater larval mass (SEL)]. RNA-seq was conducted on 5th instar BSF larvae (n = 36), representing equal proportions of the NS (F2 = 9; F3 = 9) and SEL (F2 = 9; F3 = 9) groups. A multidimensional scaling plot revealed greater gene expression variability within the NS and F2 subgroups, while the SEL group clustered separately with lower levels of variation. Comparisons between generations revealed 898 differentially expressed genes (DEGs; FDR-corrected p < 0.05), while between selection strategies, 213 DEGs were observed (FDR-corrected p < 0.05). Enrichment analyses revealed that metabolic, developmental, and defence response processes were over-expressed in the comparison between F2 and F3 larvae, while metabolic processes were the main differentiating factor between NS and SEL lines. This illustrates the functional adaptations that occur in BSF colonies across generations due to mass rearing; as well as highlighting genic dynamics associated with artificial selection for production traits that might inform future selective breeding strategies.
Collapse
Affiliation(s)
- Kelvin L Hull
- Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | | | - Melissa Lloyd
- Research and Development Department, Insect Technology Group Holdings UK Ltd., Guildford, UK
| | | | - Clint Rhode
- Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
23
|
Zhang J, Li S, Luo X, Zhang C. Emerging role of hypothalamus in the metabolic regulation in the offspring of maternal obesity. Front Nutr 2023; 10:1094616. [PMID: 36819678 PMCID: PMC9928869 DOI: 10.3389/fnut.2023.1094616] [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: 11/10/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Maternal obesity has a significant impact on the metabolism of offspring both in childhood and adulthood. The metabolic regulation of offspring is influenced by the intrauterine metabolic programming induced by maternal obesity. Nevertheless, the precise mechanisms remain unclear. The hypothalamus is the primary target of metabolic programming and the principal regulatory center of energy metabolism. Accumulating evidence has indicated the crucial role of hypothalamic regulation in the metabolism of offspring exposed to maternal obesity. This article reviews the development of hypothalamus, the role of the hypothalamic regulations in energy homeostasis, possible mechanisms underlying the developmental programming of energy metabolism in offspring, and the potential therapeutic approaches for preventing metabolic diseases later in life. Lastly, we discuss the challenges and future directions of hypothalamic regulation in the metabolism of children born to obese mothers.
Collapse
|
24
|
Bhal S, Kundu CN. Targeting crosstalk of signaling pathways in cancer stem cells: a promising approach for development of novel anti-cancer therapeutics. Med Oncol 2023; 40:82. [PMID: 36662310 DOI: 10.1007/s12032-022-01905-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/21/2022] [Indexed: 01/21/2023]
Abstract
Wnt, Hedgehog (Hh), and Notch signaling pathways are the evolutionarily conserved signaling pathways that regulate the embryonic development and also play crucial role in maintaining stemness properties of cancer stem cells (CSCs) and inducing epithelial-to-mesenchymal transition (EMT), metastasis, and angiogenesis. It has been highly challenging to inhibit the CSCs growth and proliferation as these are capable of evading chemotherapeutic drugs and cause cancer recurrence through multiple signaling pathways. Therefore, novel therapeutic strategies to target the key players involved in the crosstalk of these signaling pathways need to be developed. In this review, we have identified the interacting molecules of Wnt, Hh, and Notch pathways responsible for enhancing the malignant properties of CSCs. Analyzing the functions of these crosstalk molecules will help us to find an approach toward the development of new anti-cancer drugs for inhibition of CSCs growth and progression. Long non-coding RNAs (LncRNAs) play a significant role in various cellular processes, like chromatin remodeling, epigenetic modifications, transcriptional, and post-transcriptional regulations. Here, we have highlighted the research findings suggesting the involvement of LncRNAs in maintenance of the stemness properties of CSCs through modulation of the above-mentioned signaling pathways. We have also discussed about the different therapeutic approaches targeting those key players responsible for mediating the crosstalk between the pathways. Overall, this review article will surely help the cancer biologists to design novel anti-CSCs agents that will open up a new horizon in the field of anti-cancer therapeutics.
Collapse
Affiliation(s)
- Subhasmita Bhal
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
25
|
Chen D, Forghany Z, Liu X, Wang H, Merks RMH, Baker DA. A new model of Notch signalling: Control of Notch receptor cis-inhibition via Notch ligand dimers. PLoS Comput Biol 2023; 19:e1010169. [PMID: 36668673 PMCID: PMC9891537 DOI: 10.1371/journal.pcbi.1010169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 02/01/2023] [Accepted: 12/30/2022] [Indexed: 01/22/2023] Open
Abstract
All tissue development and replenishment relies upon the breaking of symmetries leading to the morphological and operational differentiation of progenitor cells into more specialized cells. One of the main engines driving this process is the Notch signal transduction pathway, a ubiquitous signalling system found in the vast majority of metazoan cell types characterized to date. Broadly speaking, Notch receptor activity is governed by a balance between two processes: 1) intercellular Notch transactivation triggered via interactions between receptors and ligands expressed in neighbouring cells; 2) intracellular cis inhibition caused by ligands binding to receptors within the same cell. Additionally, recent reports have also unveiled evidence of cis activation. Whilst context-dependent Notch receptor clustering has been hypothesized, to date, Notch signalling has been assumed to involve an interplay between receptor and ligand monomers. In this study, we demonstrate biochemically, through a mutational analysis of DLL4, both in vitro and in tissue culture cells, that Notch ligands can efficiently self-associate. We found that the membrane proximal EGF-like repeat of DLL4 was necessary and sufficient to promote oligomerization/dimerization. Mechanistically, our experimental evidence supports the view that DLL4 ligand dimerization is specifically required for cis-inhibition of Notch receptor activity. To further substantiate these findings, we have adapted and extended existing ordinary differential equation-based models of Notch signalling to take account of the ligand dimerization-dependent cis-inhibition reported here. Our new model faithfully recapitulates our experimental data and improves predictions based upon published data. Collectively, our work favours a model in which net output following Notch receptor/ligand binding results from ligand monomer-driven Notch receptor transactivation (and cis activation) counterposed by ligand dimer-mediated cis-inhibition.
Collapse
Affiliation(s)
- Daipeng Chen
- School of Mathematics and Statistics, Xi’an Jiaotong University, Xi’an, China
- Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - Zary Forghany
- Leiden University Medical Center (LUMC), Department of Cell & Chemical Biology, Leiden, The Netherlands
| | - Xinxin Liu
- Leiden University Medical Center (LUMC), Department of Cell & Chemical Biology, Leiden, The Netherlands
| | - Haijiang Wang
- Leiden University Medical Center (LUMC), Department of Cell & Chemical Biology, Leiden, The Netherlands
- Department of General Surgery, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Roeland M. H. Merks
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
- * E-mail: (RMHM); (DAB)
| | - David A. Baker
- Leiden University Medical Center (LUMC), Department of Cell & Chemical Biology, Leiden, The Netherlands
- * E-mail: (RMHM); (DAB)
| |
Collapse
|
26
|
Yu K, Liu Z, Cheng H, Li S, Zhang Q, Liu J, Ju HQ, Zuo Z, Zhao Q, Kang S, Liu ZX. dSCOPE: a software to detect sequences critical for liquid-liquid phase separation. Brief Bioinform 2023; 24:6927233. [PMID: 36528388 DOI: 10.1093/bib/bbac550] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/26/2022] [Accepted: 11/12/2022] [Indexed: 12/23/2022] Open
Abstract
Membrane-based cells are the fundamental structural and functional units of organisms, while evidences demonstrate that liquid-liquid phase separation (LLPS) is associated with the formation of membraneless organelles, such as P-bodies, nucleoli and stress granules. Many studies have been undertaken to explore the functions of protein phase separation (PS), but these studies lacked an effective tool to identify the sequence segments that critical for LLPS. In this study, we presented a novel software called dSCOPE (http://dscope.omicsbio.info) to predict the PS-driving regions. To develop the predictor, we curated experimentally identified sequence segments that can drive LLPS from published literature. Then sliding sequence window based physiological, biochemical, structural and coding features were integrated by random forest algorithm to perform prediction. Through rigorous evaluation, dSCOPE was demonstrated to achieve satisfactory performance. Furthermore, large-scale analysis of human proteome based on dSCOPE showed that the predicted PS-driving regions enriched various protein post-translational modifications and cancer mutations, and the proteins which contain predicted PS-driving regions enriched critical cellular signaling pathways. Taken together, dSCOPE precisely predicted the protein sequence segments critical for LLPS, with various helpful information visualized in the webserver to facilitate LLPS-related research.
Collapse
Affiliation(s)
- Kai Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zekun Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Haoyang Cheng
- Department of Computer Science, The University of Hong Kong, Hong Kong 999077, China
| | - Shihua Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Qingfeng Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jia Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhixiang Zuo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Qi Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shiyang Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ze-Xian Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
27
|
Du Y, Gao H, He C, Xin S, Wang B, Zhang S, Gong F, Yu X, Pan L, Sun F, Wang W, Xu J. An update on the biological characteristics and functions of tuft cells in the gut. Front Cell Dev Biol 2023; 10:1102978. [PMID: 36704202 PMCID: PMC9872863 DOI: 10.3389/fcell.2022.1102978] [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: 11/23/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023] Open
Abstract
The intestine is a powerful digestive system and one of the most sophisticated immunological organs. Evidence shows that tuft cells (TCs), a kind of epithelial cell with distinct morphological characteristics, play a significant role in various physiological processes. TCs can be broadly categorized into different subtypes depending on different molecular criteria. In this review, we discuss its biological properties and role in maintaining homeostasis in the gastrointestinal tract. We also emphasize its relevance to the immune system and highlight its powerful influence on intestinal diseases, including inflammations and tumors. In addition, we provide fresh insights into future clinical diagnostic and therapeutic strategies related to TCs.
Collapse
Affiliation(s)
- Yixuan Du
- Department of Oral Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Boya Wang
- Undergraduate Student of 2018 Eight Program of Clinical Medicine, Peking University People’s Hospital, Beijing, China
| | - Sitian Zhang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fengrong Gong
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinyi Yu
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Luming Pan
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fanglin Sun
- Department of Laboratory Animal Research, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Wen Wang
- Department of Laboratory Animal Research, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,*Correspondence: Jingdong Xu,
| |
Collapse
|
28
|
Reprogramming of cancer-associated fibroblasts by apoptotic cancer cells inhibits lung metastasis via Notch1-WISP-1 signaling. Cell Mol Immunol 2022; 19:1373-1391. [PMID: 36241874 PMCID: PMC9708692 DOI: 10.1038/s41423-022-00930-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/14/2022] [Indexed: 12/05/2022] Open
Abstract
The interplay between apoptotic cancer cells and the tumor microenvironment modulates cancer progression and metastasis. Cancer-associated fibroblasts (CAFs) play a crucial role in promoting these events through paracrine communication. Here, we demonstrate that conditioned medium (CM) from lung CAFs exposed to apoptotic cancer cells suppresses TGF-β1-induced migration and invasion of cancer cells and CAFs. Direct exposure of CAFs to apoptotic 344SQ cells (ApoSQ) inhibited CAF migration and invasion and the expression of CAF activation markers. Enhanced secretion of Wnt-induced signaling protein 1 (WISP-1) by CAFs exposed to ApoSQ was required for these antimigratory and anti-invasive effects. Pharmacological inhibition of Notch1 activation or siRNA-mediated Notch1 silencing prevented WISP-1 production by CAFs and reversed the antimigratory and anti-invasive effects. Enhanced expression of the Notch ligand delta-like protein 1 on the surface of ultraviolet-irradiated apoptotic lung cancer cells triggered Notch1-WISP-1 signaling. Phosphatidylserine receptor brain-specific angiogenesis inhibitor 1 (BAI1)-Rac1 signaling, which facilitated efferocytosis by CAFs, participated in crosstalk with Notch1 signaling for optimal production of WISP-1. In addition, a single injection of ApoSQ enhanced WISP-1 production, suppressed the expression of CAF activation markers in isolated Thy1+ CAFs, and inhibited lung metastasis in syngeneic immunocompetent mice via Notch1 signaling. Treatment with CM from CAFs exposed to ApoSQ suppressed tumor growth and lung metastasis, whereas treatment with WISP-1-immunodepleted CM from CAFs exposed to ApoSQ reversed the antitumorigenic and antimetastatic effects. Therefore, treatment with CM from CAFs exposed to apoptotic lung cancer cells could be therapeutically applied to suppress CAF activation, thereby preventing cancer progression and metastasis.
Collapse
|
29
|
Li J, Li J, Wang H, Chen Y, Qin J, Zeng H, Wang K, Wang S. Microscopic Raman illustrating antitumor enhancement effects by the combination drugs of γ-secretase inhibitor and cisplatin on osteosarcoma cells. JOURNAL OF BIOPHOTONICS 2022; 15:e202200189. [PMID: 36057844 DOI: 10.1002/jbio.202200189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/02/2022] [Accepted: 08/31/2022] [Indexed: 06/15/2023]
Abstract
By using Raman microspectroscopy, it aims to elucidate the cellular variations caused by the combination drug of γ-secretase inhibitor (DAPT) and cisplatin in osteosarcoma (OS) cells. Illustrated by the obtained results of spectral analysis, the intracellular composition significantly changed after combined drug actions compared to the solo DAPT treatment, indicating the synergistic effect of DAPT combined with cisplatin on OS cells. Meanwhile, multivariate curve resolution-alternating least squares (MCR-ALS) algorithm was utilized to address the biochemical constitution changes in all investigated groups including the untreated (UT), DAPT (40D) and combined drug (40D + 20C) treated cells. K-means cluster and univariate imaging were both utilized to visualize the changes in subcellular morphology and biochemical distribution. The presented study provides a unique understanding on the cellular responses to DAPT combined with cisplatin from the natural biochemical perspectives, and laids an experimental foundation for exploring the therapeutic strategies of other combined anticancer drugs in cancer cell model.
Collapse
Affiliation(s)
- Jie Li
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
- School of Physics and Electronic Engineering, Xianyang Normal University, Xianyang, Shaanxi, China
| | - Jing Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haifeng Wang
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Yishen Chen
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Jie Qin
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haishan Zeng
- Imaging Unit-Integrative Oncology Department, BC Cancer Research Center, Vancouver, Canada
| | - Kaige Wang
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Shuang Wang
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
30
|
Manni W, Min W. Signaling pathways in the regulation of cancer stem cells and associated targeted therapy. MedComm (Beijing) 2022; 3:e176. [PMID: 36226253 PMCID: PMC9534377 DOI: 10.1002/mco2.176] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/07/2022] Open
Abstract
Cancer stem cells (CSCs) are defined as a subpopulation of malignant tumor cells with selective capacities for tumor initiation, self-renewal, metastasis, and unlimited growth into bulks, which are believed as a major cause of progressive tumor phenotypes, including recurrence, metastasis, and treatment failure. A number of signaling pathways are involved in the maintenance of stem cell properties and survival of CSCs, including well-established intrinsic pathways, such as the Notch, Wnt, and Hedgehog signaling, and extrinsic pathways, such as the vascular microenvironment and tumor-associated immune cells. There is also intricate crosstalk between these signal cascades and other oncogenic pathways. Thus, targeting pathway molecules that regulate CSCs provides a new option for the treatment of therapy-resistant or -refractory tumors. These treatments include small molecule inhibitors, monoclonal antibodies that target key signaling in CSCs, as well as CSC-directed immunotherapies that harness the immune systems to target CSCs. This review aims to provide an overview of the regulating networks and their immune interactions involved in CSC development. We also address the update on the development of CSC-directed therapeutics, with a special focus on those with application approval or under clinical evaluation.
Collapse
Affiliation(s)
- Wang Manni
- Department of Biotherapy, Cancer Center, West China HospitalSichuan UniversityChengduP. R. China
| | - Wu Min
- Department of Biomedical Sciences, School of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| |
Collapse
|
31
|
Saini N, Bheeshmachar G, Sarin A. Sirtuin1 meditated modification of Notch1 intracellular domain regulates nucleolar localization and activation of distinct signaling cascades. Front Cell Dev Biol 2022; 10:988816. [PMID: 36211456 PMCID: PMC9539544 DOI: 10.3389/fcell.2022.988816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Notch signaling is involved in cell fate decisions in the development and maintenance of tissue homeostasis. Spatial regulation of the Notch1 intracellular domain (NIC1), has been shown to underpin signaling outcomes mediated by this receptor. We recently reported a putative Nucleolar Localization Sequence (NoLS) in NIC1. Here we investigate if the putative NoLS identified in NIC1 regulates localization in the nucleolus and anti-apoptotic activity. Confocal imaging of live cells expressing NIC1 or forms modified by deletion or site-directed mutagenesis established that the putative NoLS in NIC1 is required for nucleolar localization and regulated by the deacetylase Sirtuin1. Subsequent analysis of anti-apoptotic activity revealed signaling cascades linked to nucleolar localization. For this, etoposide and 4-Nitroquinoline 1-oxide, an inhibitor of topoisomerase-II and a UV mimetic drug respectively, were used as prototypic triggers of genomic damage in a mammalian cell line. While NIC1 blocked apoptosis regardless of its localization to the nucleoplasm or nucleolus, modifications of NIC1 which promoted localization to the nucleolus triggered a dependence on the nucleolar proteins fibrillarin and nucleolin for anti-apoptotic activity. Further, cells co-expressing NIC1 and Sirtuin1 (but not its catalytically inactive form), confirmed both spatial regulation and the switch to dependence on the nucleolar proteins. Finally, site-directed mutagenesis showed that the NoLS lysine residues are targets of Sirtuin1 activity. NIC1 mediated transcription is not similarly regulated. Thus, NIC1 localization to the nucleolus is regulated by Sirtuin1 modification of the lysine residues in NoLS and triggers a distinct signaling cascade involving nucleolar intermediates for anti-apoptotic activity.
Collapse
|
32
|
Ray A, Li X. A Notch-dependent transcriptional mechanism controls expression of temporal patterning factors in Drosophila medulla. eLife 2022; 11:e75879. [PMID: 36040415 PMCID: PMC9427115 DOI: 10.7554/elife.75879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 07/19/2022] [Indexed: 11/24/2022] Open
Abstract
Temporal patterning is an important mechanism for generating a great diversity of neuron subtypes from a seemingly homogenous progenitor pool in both vertebrates and invertebrates. Drosophila neuroblasts are temporally patterned by sequentially expressed Temporal Transcription Factors (TTFs). These TTFs are proposed to form a transcriptional cascade based on mutant phenotypes, although direct transcriptional regulation between TTFs has not been verified in most cases. Furthermore, it is not known how the temporal transitions are coupled with the generation of the appropriate number of neurons at each stage. We use neuroblasts of the Drosophila optic lobe medulla to address these questions and show that the expression of TTFs Sloppy-paired 1/2 (Slp1/2) is directly regulated at the transcriptional level by two other TTFs and the cell-cycle dependent Notch signaling through two cis-regulatory elements. We also show that supplying constitutively active Notch can rescue the delayed transition into the Slp stage in cell cycle arrested neuroblasts. Our findings reveal a novel Notch-pathway dependent mechanism through which the cell cycle progression regulates the timing of a temporal transition within a TTF transcriptional cascade.
Collapse
Affiliation(s)
- Alokananda Ray
- Department of Cell and Developmental Biology, University of Illinois at Urbana-ChampaignUrbanaUnited States
| | - Xin Li
- Department of Cell and Developmental Biology, University of Illinois at Urbana-ChampaignUrbanaUnited States
| |
Collapse
|
33
|
Bioinformatics analysis and experimental verification of Notch signalling pathway-related miRNA-mRNA subnetwork in extracellular vesicles during Echinococcus granulosus encystation. Parasit Vectors 2022; 15:272. [PMID: 35906657 PMCID: PMC9338502 DOI: 10.1186/s13071-022-05391-8] [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: 04/28/2022] [Accepted: 07/05/2022] [Indexed: 12/02/2022] Open
Abstract
Background Encystation of the protoscoleces (PSCs) of Echinococcus granulosus is the main cause of secondary hydatid dissemination in the intermediate host. Extracellular vesicles (EVs) can transfer miRNAs into parasite cells to regulate mRNA expression. However, loading of developmental pathway-related miRNAs, such as those related to the Notch signalling pathway in EVs is unclear. Thus, we screened the miRNA-mRNA subnetwork involved in the Notch pathway during E. granulosus encystation in vitro and assessed changes in expression in the parasite and EVs. Methods mRNAs and miRNAs differentially expressed (DE) between PSCs and microcysts (MCs) were screened using high-throughput sequencing. DE mRNAs obtained from transcriptome analysis were intersected with mRNAs predicted to be targets of the conserved DE miRNAs of a small RNA library. DE miRNA functions were analysed using public databases, and a miRNA–mRNA subnetwork related to the Notch pathway was established. Notch pathway-related mRNA and miRNA expression of worms and EVs at different times was verified. Results In total, 1445 DE mRNAs between MCs and PSCs were screened after the intersection between 1586 DE mRNAs from the transcriptome and 9439 target mRNAs predicted using 39 DE miRNAs from the small RNA library. The DE mRNAs were clustered into 94 metabolic pathways, including the Notch pathway. Five DE miRNAs, including the most significantly expressed new DE miRNA, egr-new-mir0694-3p, corresponding to four target mRNAs (EgrG_000892700, EgrG_001029400, EgrG_001081400 and EgrG_000465800) were all enriched in the Notch pathway. The expression of the above mRNAs and miRNAs was consistent with the results of high-throughput sequencing, and the expression of each miRNA in EVs was verified. Annotated as ADAM17/TACE in the Notch pathway, EgrG_000892700 was down-regulated during PSC encystation. egr-miR-4989-3p and egr-miR-277a-3p expression in EVs after encystation was nearly five times that in EVs before encystation, which might regulate the expression of EgrG_000892700. Conclusions Five miRNAs corresponding to four target mRNAs may be involved in regulating the Notch pathway during the PSC encystation. EVs may regulate the expression of EgrG_000892700 in PSCs because of continuous targeting of egr-miR-4989-3p and egr-miR-277a-3p and participate in the regulation the Notch pathway. The study might expand new ideas for blocking the secondary infection of E. granulosus PSCs via EVs miRNAs. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05391-8.
Collapse
|
34
|
Liu Z, Li W, Cao Y, Zhang X, Yang K, Yin F, Yang M, Peng P. Effects of the interaction of Notch and TLR4 pathways on inflammation and heart function in septic heart. Open Life Sci 2022; 17:744-755. [PMID: 35891967 PMCID: PMC9281592 DOI: 10.1515/biol-2022-0076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 03/22/2022] [Accepted: 04/07/2022] [Indexed: 11/15/2022] Open
Abstract
We investigated the role of the interaction between the Notch and Toll-like receptor 4 (TLR4) pathways in septic myocardial injury. The sepsis model was induced in rats with lipopolysaccharide (LPS). Rats were divided into control, LPS, LPS + TAK242 ((6R)-6-[N-(2-chloro-4-fluorophenyl)sulfamoyl]cyclohex-1-ene-1-carboxylate) and LPS + DAPT (N-[N-(3,5-difluorophenacetyl)-l-alanyl]-s-phenylglycinetbutylester) groups. Heart function was evaluated with a Cardiac Doppler ultrasound. Myocardial morphological changes were detected by hematoxylin-eosin staining (H&E). Apoptosis was assessed by a TUNEL assay. The mRNA and protein levels were detected with real-time PCR, Western blot, and immunohistochemistry analysis. We found that heart function in the LPS + TAK242 group was significantly improved, but not in the LPS + DAPT group. LPS + TAK242 had a lower level of degeneration and necrosis of cardiomyocytes and inflammatory cell infiltration, as well as lower apoptosis and caspase-3 expression than the LPS group. Compared with the LPS group, the inflammatory cell infiltration was reduced in the LPS + DAPT group, while the degeneration and necrosis of cardiomyocytes were not obviously improved. Additionally, the expression levels of tumor necrosis factor-α and Interleukin-6, the protein contents of Notch intracellular domain and Hes1, and the P65 nuclear factor kappa-B (NF-κB) to P-P65 NF-κB ratio in LPS + TAK242 group and LPS + DAPT group were significantly lower than those in LPS group. Conclusively, the interaction between TLR4 and Notch signaling pathways enhances the inflammatory response in the septic heart by activating NF-κB. Blocking the TLR4 pathway with TAK242 can improve heart dysfunction and myocardial damage in sepsis, while blocking the Notch pathway with DAPT cannot effectively prevent heart dysfunction and myocardial damage in sepsis.
Collapse
Affiliation(s)
- Ziyang Liu
- Intensive Care Unit, Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Wenli Li
- Emergency Department of Internal Medicine, Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Yang Cao
- Intensive Care Unit, Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Xiaoxia Zhang
- Intensive Care Unit, Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Kai Yang
- Intensive Care Unit, Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Fukang Yin
- Intensive Care Unit, Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Meng Yang
- Intensive Care Unit, Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Peng Peng
- Intensive Care Unit, Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| |
Collapse
|
35
|
Mugisha S, Di X, Disoma C, Jiang H, Zhang S. Fringe family genes and their modulation of Notch signaling in cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188746. [PMID: 35660646 DOI: 10.1016/j.bbcan.2022.188746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 11/26/2022]
Abstract
Fringes are glycosyltransferases that transfer N-acetylglucosamine to the O-linked fucose of Notch receptors. They regulate the Notch signaling activity that drives tumor formation and progression, resulting in poor prognosis. However, the specific tumor-promoting role of Fringes differs depending on the type of cancer. Although a particular Fringe member could act as a tumor suppressor in one cancer type, it may act as an oncogene in another. This review discusses the tumorigenic role of the Fringe family (lunatic fringe, manic fringe, and radical fringe) in modulating Notch signaling in various cancers. Although the crucial functions of Fringes continue to emerge as more mechanistic studies are being pursued, further translational research is needed to explore their roles and therapeutic benefits in various malignancies.
Collapse
Affiliation(s)
- Samson Mugisha
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Xiaotang Di
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Cyrollah Disoma
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Hao Jiang
- Department of Biomedical Informatics, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China.
| | - Shubing Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan 410013, PR China.
| |
Collapse
|
36
|
Wang Y, Xie W, Feng Y, Xu Z, He Y, Xiong Y, Chen L, Li X, Liu J, Liu G, Wu Q. Epithelial‑derived exosomes promote M2 macrophage polarization via Notch2/SOCS1 during mechanical ventilation. Int J Mol Med 2022; 50:96. [PMID: 35616134 PMCID: PMC9170191 DOI: 10.3892/ijmm.2022.5152] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022] Open
Abstract
Alveolar macrophages (AMs) play an essential role in ventilator-induced lung injury (VILI). Exosomes and their cargo, including microRNAs (miRNAs/miRs) serve as regulators of the intercellular communications between macrophages and epithelial cells (ECs), and are involved in maintaining homeostasis in lung tissue. The present study found that exosomes released by ECs subjected to cyclic stretching promoted M2 macrophage polarization. It was demonstrated that miR-21a-5p, upregulated in epithelial-derived exosomes, increased the percentage of M2 macrophages by suppressing the expression of Notch2 and the suppressor of cytokine signaling 1 (SOCS1). The overexpression of Notch2 decreased the percentage of M2 macrophages. However, these effects were reversed by the downregulation of SOCS1. The percentage of M2 macrophages was increased in both short-term high- and low-tidal-volume mechanical ventilation, and the administration of exosomes-derived from cyclically stretched ECs had the same function. However, the administration of miR-21a-5p antagomir decreased M2 macrophage activation induced by cyclically stretched ECs or ventilation. Thus, the present study demonstrates that the intercellular transferring of exosomes from ECs to AMs promotes M2 macrophage polarization. Exosomes may prove to be a novel treatment for VILI.
Collapse
Affiliation(s)
- Yanting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wanli Xie
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yiqi Feng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhenzhen Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yuyao He
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yue Xiong
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lu Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xia Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jie Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Guoyang Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qingping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
37
|
Garg C, khan H, Kaur A, Singh TG, Sharma VK, Singh SK. Therapeutic Implications of Sonic Hedgehog Pathway in Metabolic Disorders: Novel Target for Effective Treatment. Pharmacol Res 2022; 179:106194. [DOI: 10.1016/j.phrs.2022.106194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 12/13/2022]
|
38
|
Ossola C, Kalebic N. Roots of the Malformations of Cortical Development in the Cell Biology of Neural Progenitor Cells. Front Neurosci 2022; 15:817218. [PMID: 35069108 PMCID: PMC8766818 DOI: 10.3389/fnins.2021.817218] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022] Open
Abstract
The cerebral cortex is a structure that underlies various brain functions, including cognition and language. Mammalian cerebral cortex starts developing during the embryonic period with the neural progenitor cells generating neurons. Newborn neurons migrate along progenitors’ radial processes from the site of their origin in the germinal zones to the cortical plate, where they mature and integrate in the forming circuitry. Cell biological features of neural progenitors, such as the location and timing of their mitoses, together with their characteristic morphologies, can directly or indirectly regulate the abundance and the identity of their neuronal progeny. Alterations in the complex and delicate process of cerebral cortex development can lead to malformations of cortical development (MCDs). They include various structural abnormalities that affect the size, thickness and/or folding pattern of the developing cortex. Their clinical manifestations can entail a neurodevelopmental disorder, such as epilepsy, developmental delay, intellectual disability, or autism spectrum disorder. The recent advancements of molecular and neuroimaging techniques, along with the development of appropriate in vitro and in vivo model systems, have enabled the assessment of the genetic and environmental causes of MCDs. Here we broadly review the cell biological characteristics of neural progenitor cells and focus on those features whose perturbations have been linked to MCDs.
Collapse
|
39
|
Epigenetic Modulation of Gremlin-1/NOTCH Pathway in Experimental Crescentic Immune-Mediated Glomerulonephritis. Pharmaceuticals (Basel) 2022; 15:ph15020121. [PMID: 35215234 PMCID: PMC8876310 DOI: 10.3390/ph15020121] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/20/2022] Open
Abstract
Crescentic glomerulonephritis is a devastating autoimmune disease that without early and properly treatment may rapidly progress to end-stage renal disease and death. Current immunosuppressive treatment provides limited efficacy and an important burden of adverse events. Epigenetic drugs are a source of novel therapeutic tools. Among them, bromodomain and extraterminal domain (BET) inhibitors (iBETs) block the interaction between bromodomains and acetylated proteins, including histones and transcription factors. iBETs have demonstrated protective effects on malignancy, inflammatory disorders and experimental kidney disease. Recently, Gremlin-1 was proposed as a urinary biomarker of disease progression in human anti-neutrophil cytoplasmic antibody (ANCA)-associated crescentic glomerulonephritis. We have now evaluated whether iBETs could regulate Gremlin-1 in experimental anti-glomerular basement membrane nephritis induced by nephrotoxic serum (NTS) in mice, a model resembling human crescentic glomerulonephritis. In NTS-injected mice, the iBET JQ1 inhibited renal Gremlin-1 overexpression and diminished glomerular damage, restoring podocyte numbers. Chromatin immunoprecipitation assay demonstrated BRD4 enrichment of the Grem-1 gene promoter in injured kidneys, consistent with Gremlin-1 epigenetic regulation. Moreover, JQ1 blocked BRD4 binding and inhibited Grem-1 gene transcription. The beneficial effect of iBETs was also mediated by modulation of NOTCH pathway. JQ1 inhibited the gene expression of the NOTCH effectors Hes-1 and Hey-1 in NTS-injured kidneys. Our results further support the role for epigenetic drugs, such as iBETs, in the treatment of rapidly progressive crescentic glomerulonephritis.
Collapse
|
40
|
Dahmardeh Ghalehno A, Boustan A, Abdi H, Aganj Z, Mosaffa F, Jamialahmadi K. The Potential for Natural Products to Overcome Cancer Drug Resistance by Modulation of Epithelial-Mesenchymal Transition. Nutr Cancer 2022; 74:2686-2712. [PMID: 34994266 DOI: 10.1080/01635581.2021.2022169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The acquisition of resistance and ultimately disease relapse after initial response to chemotherapy put obstacles in the way of cancer therapy. Epithelial-mesenchymal transition (EMT) is a biologic process that epithelial cells alter to mesenchymal cells and acquire fibroblast-like properties. EMT plays a significant role in cancer metastasis, motility, and survival. Recently, emerging evidence suggested that EMT pathways are very important in making drug-resistant involved in cancer. Natural products are gradually emerging as a valuable source of safe and effective anticancer compounds. Natural products could interfere with the different processes implicated in cancer drug resistance by reversing the EMT process. In this review, we illustrate the molecular mechanisms of EMT in the emergence of cancer metastasis. We then present the role of natural compounds in the suppression of EMT pathways in different cancers to overcome cancer cell drug resistance and improve tumor chemotherapy. HighlightsDrug-resistance is one of the obstacles to cancer treatment.EMT signaling pathways have been correlated to tumor invasion, metastasis, and drug-resistance.Various studies on the relationship between EMT and resistance to chemotherapy agents were reviewed.Different anticancer natural products with EMT inhibitory properties and drug resistance reversal effects were compared.
Collapse
Affiliation(s)
- Asefeh Dahmardeh Ghalehno
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arad Boustan
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hakimeh Abdi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Aganj
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
41
|
Sharma V, Sachan N, Mutsuddi M, Mukherjee A. Somatic Clonal Analyses Using FLP/FRT and MARCM System to Understand Notch Signaling Mechanism and Its Regulation. Methods Mol Biol 2022; 2472:83-94. [PMID: 35674894 DOI: 10.1007/978-1-0716-2201-8_8] [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: 06/15/2023]
Abstract
Notch signaling regulates an array of developmental decisions and has been implicated in a multitude of diseases, including cancer over the past a few decades. The simplicity and versatility of the Notch pathway in Drosophila make it an ardent system to study Notch biology, its regulation, and functions. In this chapter, we highlight the use of two powerful techniques, namely, FLP/FRT and MARCM in the study of Notch signaling. These mosaic analysis techniques are powerful tools to analyze gene functions in different biological processes. The section briefly explains the principle and the protocols with suitable examples.
Collapse
Affiliation(s)
- Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Nalani Sachan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
42
|
Nueda ML, Baladrón V. Mammalian NOTCH Receptor Activation and Signaling Protocols. Methods Mol Biol 2022; 2472:67-82. [PMID: 35674893 DOI: 10.1007/978-1-0716-2201-8_7] [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: 06/15/2023]
Abstract
The NOTCH signaling pathway is one of the highly conserved key pathways involved in most cell differentiation and proliferation processes during both developmental and adult stages in most animals. The NOTCH signaling pathway appears to be very simple but the existence of several receptors and ligands, their posttranslational modifications, their activation in the cell surface and its migration to the cell nucleus, as well as their interaction with multiple signaling pathways in the cytoplasm and the nucleus of cells, make the study of its function very complex.To determine the activation of NOTCH signaling in animal cells, several complementary approaches can be performed. One of them is the analysis of the transcription of NOTCH receptor target genes HES/HEY by qRT-PCR and Western blot. This approach would give us an idea of the global NOTCH activation and signaling. We can also analyze the NOTCH transcriptional activity by luciferase assays to determine the global or specific activation of NOTCH receptors under a given treatment or in response to the modification of gene expression. On the other hand, we can determine the specific activation of each NOTCH receptor by Western blot with antibodies that recognize the active forms of each NOTCH receptor. For this assay will be very important to collect the cells to be analyzed under the appropriate conditions. Finally, we can detect the intracellular domain of each NOTCH receptor into the cell nucleus by confocal microscopy using the appropriate antibodies that recognize the intracellular domain of the receptors.
Collapse
Affiliation(s)
- María-Luisa Nueda
- Biochemistry and Molecular Biology Branch, School of Pharmacy/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-LaMancha/CSIC, Albacete, Spain.
| | - Victoriano Baladrón
- Biochemistry and Molecular Biology Branch, Albacete Medical School/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-LaMancha/CSIC, Albacete, Spain.
| |
Collapse
|
43
|
Zhang T, Liu X, Su K, Zheng Q, Liu P, Xu Z, Zhang Y. A novel mechanism of the lncRNA PTTG3P/miR-142-5p/JAG1 axis modulating tongue cancer cell phenotypes through the Notch1 signaling. Cells Dev 2021; 169:203762. [PMID: 34952204 DOI: 10.1016/j.cdev.2021.203762] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/11/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022]
Abstract
Tongue cancer is the most prevalent type of oral cancer. Our previous study revealed that JAG1 exerted an oncogenic effect on tongue carcinoma through the JAG1/Notch pathway. In this study, a lncRNA PTTG3P which was upregulated in tongue cancer, was found to be positively correlated with JAG1. In CAL-27 and SCC4 cells, PTTG3P silencing significantly decreased JAG1 proteins and the ability of tongue tumor cells to proliferate and migrate. PTTG3P overexpression exhibited the opposite effect on CAL-27 and SCC4 cells. PPTG3P directly bound miR-142-5p, and miR-142-5p directly bound 3'UTR of JAG1 and inhibited the expression levels of JAG1. As opposed to PTTG3P silencing, miR-142-5p inhibition increased JAG1 protein levels and tongue cancer cell proliferation and migration; moreover, miR-142-5p inhibition substantially reversed the effects of PTTG3P silencing. Finally, the PPTG3P/miR-142-5p axis regulated the level of NICD, Notch downstream c-myc, and cyclin D1, as well as EMT markers Snail, Twist, and Vimentin. In conclusion, the PTTG3P/miR-142-5p axis modulates tongue cancer aggressiveness through JAG1, potentially through a JAG1/Notch signaling pathway.
Collapse
Affiliation(s)
- Tonghan Zhang
- Department of Oral and Maxillofacial Surgery, Affiliated Zhongshan Hospital, Sun Yat-sen University, Zhongshan, Guangdong 528400, China
| | - Xiaoling Liu
- Department of Oral and Maxillofacial Surgery, Affiliated Zhongshan Hospital, Sun Yat-sen University, Zhongshan, Guangdong 528400, China
| | - Kui Su
- Department of Oral and Maxillofacial Surgery, Affiliated Zhongshan Hospital, Sun Yat-sen University, Zhongshan, Guangdong 528400, China.
| | - Qiaoyi Zheng
- Department of Oral and Maxillofacial Surgery, Affiliated Zhongshan Hospital, Sun Yat-sen University, Zhongshan, Guangdong 528400, China
| | - Peng Liu
- Department of Oral and Maxillofacial Surgery, Affiliated Zhongshan Hospital, Sun Yat-sen University, Zhongshan, Guangdong 528400, China
| | - Zhijie Xu
- Department of Oral and Maxillofacial Surgery, Affiliated Zhongshan Hospital, Sun Yat-sen University, Zhongshan, Guangdong 528400, China
| | - Yonghao Zhang
- Department of Oral and Maxillofacial Surgery, Affiliated Zhongshan Hospital, Sun Yat-sen University, Zhongshan, Guangdong 528400, China
| |
Collapse
|
44
|
Cucu I, Nicolescu MI. A Synopsis of Signaling Crosstalk of Pericytes and Endothelial Cells in Salivary Gland. Dent J (Basel) 2021; 9:dj9120144. [PMID: 34940041 PMCID: PMC8700478 DOI: 10.3390/dj9120144] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
The salivary gland (SG) microvasculature constitutes a dynamic cellular organization instrumental to preserving tissue stability and homeostasis. The interplay between pericytes (PCs) and endothelial cells (ECs) culminates as a key ingredient that coordinates the development, maturation, and integrity of vessel building blocks. PCs, as a variety of mesenchymal stem cells, enthrall in the field of regenerative medicine, supporting the notion of regeneration and repair. PC-EC interconnections are pivotal in the kinetic and intricate process of angiogenesis during both embryological and post-natal development. The disruption of this complex interlinkage corresponds to SG pathogenesis, including inflammation, autoimmune disorders (Sjögren’s syndrome), and tumorigenesis. Here, we provided a global portrayal of major signaling pathways between PCs and ECs that cooperate to enhance vascular steadiness through the synergistic interchange. Additionally, we delineated how the crosstalk among molecular networks affiliate to contribute to a malignant context. Additionally, within SG microarchitecture, telocytes and myoepithelial cells assemble a labyrinthine companionship, which together with PCs appear to synchronize the regenerative potential of parenchymal constituents. By underscoring the intricacy of signaling cascades within cellular latticework, this review sketched a perceptive basis for target-selective drugs to safeguard SG function.
Collapse
Affiliation(s)
- Ioana Cucu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Mihnea Ioan Nicolescu
- Division of Histology, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Laboratory of Radiobiology, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
- Correspondence:
| |
Collapse
|
45
|
Jarosch S, Köhlen J, Sarker RS, Steiger K, Janssen KP, Christians A, Hennig C, Holler E, D'Ippolito E, Busch DH. Multiplexed imaging and automated signal quantification in formalin-fixed paraffin-embedded tissues by ChipCytometry. CELL REPORTS METHODS 2021; 1:100104. [PMID: 35475000 PMCID: PMC9017205 DOI: 10.1016/j.crmeth.2021.100104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/30/2021] [Accepted: 10/06/2021] [Indexed: 12/17/2022]
Abstract
Deciphering the spatial composition of cells in tissues is essential for detailed understanding of biological processes in health and disease. Recent technological advances enabled the assessment of the enormous complexity of tissue-derived parameters by highly multiplexed tissue imaging (HMTI), but elaborate machinery and data analyses are required. This severely limits broad applicability of HMTI. Here we demonstrate for the first time the application of ChipCytometry technology, which has unique features for widespread use, on formalin-fixed paraffin-embedded samples, the most commonly used storage technique of clinically relevant patient specimens worldwide. The excellent staining quality permits workflows for automated quantification of signal intensities, which we further optimized to compensate signal spillover from neighboring cells. In combination with the high number of validated markers, the reported platform can be used from unbiased analyses of tissue composition to detection of phenotypically complex rare cells, and can be easily implemented in both routine research and clinical pathology.
Collapse
Affiliation(s)
- Sebastian Jarosch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Jan Köhlen
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Rim S.J. Sarker
- Comparative Experimental Pathology, Institute for Pathology, Technical University of Munich, 81675 Munich, Germany
| | - Katja Steiger
- Comparative Experimental Pathology, Institute for Pathology, Technical University of Munich, 81675 Munich, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | | | | | - Ernst Holler
- Department of Hematology/Oncology, University Medical Center, 93053 Regensburg, Germany
| | - Elvira D'Ippolito
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, 81675 Munich, Germany
| |
Collapse
|
46
|
Torab P, Yan Y, Ahmed M, Yamashita H, Warrick JI, Raman JD, DeGraff DJ, Wong PK. Intratumoral Heterogeneity Promotes Collective Cancer Invasion through NOTCH1 Variation. Cells 2021; 10:3084. [PMID: 34831307 PMCID: PMC8619970 DOI: 10.3390/cells10113084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 12/20/2022] Open
Abstract
Cellular and molecular heterogeneity within tumors has long been associated with the progression of cancer to an aggressive phenotype and a poor prognosis. However, how such intratumoral heterogeneity contributes to the invasiveness of cancer is largely unknown. Here, using a tumor bioengineering approach, we investigate the interaction between molecular subtypes within bladder microtumors and the corresponding effects on their invasiveness. Our results reveal heterogeneous microtumors formed by multiple molecular subtypes possess enhanced invasiveness compared to individual cells, even when both cells are not invasive individually. To examine the molecular mechanism of intratumoral heterogeneity mediated invasiveness, live single cell biosensing, RNA interference, and CRISPR-Cas9 gene editing approaches were applied to investigate and control the composition of the microtumors. An agent-based computational model was also developed to evaluate the influence of NOTCH1 variation on DLL4 expression within a microtumor. The data indicate that intratumoral variation in NOTCH1 expression can lead to upregulation of DLL4 expression within the microtumor and enhancement of microtumor invasiveness. Overall, our results reveal a novel mechanism of heterogeneity mediated invasiveness through intratumoral variation of gene expression.
Collapse
Affiliation(s)
- Peter Torab
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USA;
| | - Yue Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; (Y.Y.); (M.A.)
| | - Mona Ahmed
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; (Y.Y.); (M.A.)
| | - Hironobu Yamashita
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (H.Y.); (J.I.W.); (D.J.D.)
| | - Joshua I. Warrick
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (H.Y.); (J.I.W.); (D.J.D.)
- Penn State Health Milton S., Hershey Medical Center, Department of Surgery, Hershey, PA 17033, USA;
| | - Jay D. Raman
- Penn State Health Milton S., Hershey Medical Center, Department of Surgery, Hershey, PA 17033, USA;
| | - David J. DeGraff
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (H.Y.); (J.I.W.); (D.J.D.)
- Penn State Health Milton S., Hershey Medical Center, Department of Surgery, Hershey, PA 17033, USA;
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Pak Kin Wong
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USA;
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; (Y.Y.); (M.A.)
- Penn State Health Milton S., Hershey Medical Center, Department of Surgery, Hershey, PA 17033, USA;
| |
Collapse
|
47
|
Bonnet C, González S, Roberts JS, Robertson SYT, Ruiz M, Zheng J, Deng SX. Human limbal epithelial stem cell regulation, bioengineering and function. Prog Retin Eye Res 2021; 85:100956. [PMID: 33676006 PMCID: PMC8428188 DOI: 10.1016/j.preteyeres.2021.100956] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/21/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
The corneal epithelium is continuously renewed by limbal stem/progenitor cells (LSCs), a cell population harbored in a highly regulated niche located at the limbus. Dysfunction and/or loss of LSCs and their niche cause limbal stem cell deficiency (LSCD), a disease that is marked by invasion of conjunctival epithelium into the cornea and results in failure of epithelial wound healing. Corneal opacity, pain, loss of vision, and blindness are the consequences of LSCD. Successful treatment of LSCD depends on accurate diagnosis and staging of the disease and requires restoration of functional LSCs and their niche. This review highlights the major advances in the identification of potential LSC biomarkers and components of the LSC niche, understanding of LSC regulation, methods and regulatory standards in bioengineering of LSCs, and diagnosis and staging of LSCD. Overall, this review presents key points for researchers and clinicians alike to consider in deepening the understanding of LSC biology and improving LSCD therapies.
Collapse
Affiliation(s)
- Clémence Bonnet
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA; Cornea Department, Paris University, Cochin Hospital, AP-HP, F-75014, Paris, France
| | - Sheyla González
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA
| | - JoAnn S Roberts
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA
| | - Sarah Y T Robertson
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA
| | - Maxime Ruiz
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA
| | - Jie Zheng
- Basic Science Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA
| | - Sophie X Deng
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
48
|
Zhang F, Chen Y, Shen J, Zhang J. The Ubiquitin Conjugating Enzyme UbcD1 is Required for Notch Signaling Activation During Drosophila Wing Development. Front Genet 2021; 12:770853. [PMID: 34712275 PMCID: PMC8546230 DOI: 10.3389/fgene.2021.770853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Notch signaling pathway plays crucial roles in animal development. Protein ubiquitination contributes to Notch signaling regulation by governing the stability and activity of major signaling components. Studies in Drosophila have identified multiple ubiquitin ligases and deubiquitinating enzymes that modify Notch ligand and receptor proteins. The fate of ubiquitinated substrates depend on topologies of the attached ubiquitin chains, which are determined by the ubiquitin conjugating enzymes (E2 enzymes). However, which E2 enzymes participate in Notch signal transduction remain elusive. Here, we report that the E2 enzyme UbcD1 is required for Notch signaling activation during Drosophila wing development. Mutations of UbcD1 lead to marginal nicks in the adult wing and reduction of Notch signaling targets expression in the wing imaginal disc. Genetic analysis reveal that UbcD1 functions in the signaling receiving cells prior to cleavage of the Notch protein. We provide further evidence suggesting that UbcD1 is likely involved in endocytic trafficking of Notch protein. Our results demonstrate that UbcD1 positively regulates Notch signaling and thus reveal a novel role of UbcD1 in development.
Collapse
Affiliation(s)
- Fengchao Zhang
- MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yao Chen
- MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jie Shen
- MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Junzheng Zhang
- MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
49
|
Huang S, Fu D, Wan Z, Li M, Li H, Chong T. Effects of a gamma-secretase inhibitor of notch signalling on transforming growth factor β1-induced urethral fibrosis. J Cell Mol Med 2021; 25:8796-8808. [PMID: 34363303 PMCID: PMC8435429 DOI: 10.1111/jcmm.16837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/10/2021] [Accepted: 07/24/2021] [Indexed: 11/30/2022] Open
Abstract
Urethral stricture (US) is a common disorder of the lower urinary tract in men caused by fibrosis. The recurrence rate of US is high; however, there are no effective therapies to prevent or treat urethral fibrosis. The pathogenesis of urethral fibrosis involves myofibroblast activation and excessive extracellular matrix (ECM) deposition. The molecular mechanisms underlying this pathological activation are not completely understood. It has been demonstrated that Notch signalling contributes to the development of fibrosis and inflammation. However, whether this contributes to urethral fibrosis remains unclear. In this study, activation of Notch signalling was observed in patients with US. Additionally, it was noted that activation of Notch signalling promoted ECM production and myofibroblast activation in human urethral scar fibroblasts (HUSFs) treated with transforming growth factor (TGF) β1. However, the Notch inhibitor N‐[N‐(3,5‐difluorophenacetyl)‐L‐alanyl]‐S‐phenylglycine t‐butyl ester (DAPT) suppressed activation of Notch signalling as well as proliferation and migration of the TGFβ1‐treated HUSFs. Additionally, DAPT ameliorated TGFβ1‐induced urethral fibrosis in Sprague Dawley rats by suppressing ECM production, myofibroblast activation and the TGFβ signalling pathway. These findings demonstrate that Notch signalling may be a promising and potential target in the prevention or treatment of urethral fibrosis.
Collapse
Affiliation(s)
- Shanlong Huang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Delai Fu
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ziyan Wan
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Min Li
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hecheng Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tie Chong
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
50
|
Edwards A, Brennan K. Notch Signalling in Breast Development and Cancer. Front Cell Dev Biol 2021; 9:692173. [PMID: 34295896 PMCID: PMC8290365 DOI: 10.3389/fcell.2021.692173] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/07/2021] [Indexed: 12/22/2022] Open
Abstract
The Notch signalling pathway is a highly conserved developmental signalling pathway, with vital roles in determining cell fate during embryonic development and tissue homeostasis. Aberrant Notch signalling has been implicated in many disease pathologies, including cancer. In this review, we will outline the mechanism and regulation of the Notch signalling pathway. We will also outline the role Notch signalling plays in normal mammary gland development and how Notch signalling is implicated in breast cancer tumorigenesis and progression. We will cover how Notch signalling controls several different hallmarks of cancer within epithelial cells with sections focussed on its roles in proliferation, apoptosis, invasion, and metastasis. We will provide evidence for Notch signalling in the breast cancer stem cell phenotype, which also has implications for therapy resistance and disease relapse in breast cancer patients. Finally, we will summarise the developments in therapeutic targeting of Notch signalling, and the pros and cons of this approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Abigail Edwards
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Keith Brennan
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|