1
|
Sergio I, Varricchio C, Squillante F, Cantale Aeo NM, Campese AF, Felli MP. Notch Inhibitors and BH3 Mimetics in T-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2024; 25:12839. [PMID: 39684550 DOI: 10.3390/ijms252312839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy with poor response to conventional therapy, derived from hematopoietic progenitors committed to T-cell lineage. Relapsed/Refractory patients account for nearly 20% of childhood and 45% of adult cases. Aberrant Notch signaling plays a critical role in T-ALL pathogenesis and therapy resistance. Notch inhibition is a promising therapeutic target for personalized medicine, and a variety of strategies to prevent Notch activation, including γ-secretase (GS) inhibitors (GSIs) and antibodies neutralizing Notch receptors or ligands, have been developed. Disruption of apoptosis is pivotal in cancer development and progression. Different reports evidenced the interplay between Notch and the anti-apoptotic Bcl-2 family proteins in T-ALL. Although based on early research data, this review discusses recent advances in directly targeting Notch receptors and the use of validated BH3 mimetics for the treatment of T-ALL and their combined action in light of current evidence of their use.
Collapse
Affiliation(s)
- Ilaria Sergio
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Claudia Varricchio
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Squillante
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | | | | | - Maria Pia Felli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
2
|
Kaveh Zenjanab M, Hashemzadeh N, Alimohammadvand S, Sharifi-Azad M, Dalir Abdolahinia E, Jahanban-Esfahlan R. Notch Signaling Suppression by Golden Phytochemicals: Potential for Cancer Therapy. Adv Pharm Bull 2024; 14:302-313. [PMID: 39206407 PMCID: PMC11347744 DOI: 10.34172/apb.2024.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/09/2024] [Accepted: 03/03/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer is one of the main causes of mortality worldwide. Cancer cells are characterized by unregulated cellular processes, including proliferation, progression, and angiogenesis. The occurrence of these processes is due to the dysregulation of various signaling pathways such as NF-κB (nuclear factor-κB), Wnt/beta-catenin, Notch signaling and MAPK (mitogen-activated protein kinases). Notch signaling pathways cause the progression of various types of malignant tumors. Among the phytochemicals for cancer therapy, several have attracted great interest, including curcumin, genistein, quercetin, silibinin, resveratrol, cucurbitacin and glycyrrhizin. Given the great cellular and molecular heterogeneity within tumors and the high toxicity and side effects of synthetic chemotherapeutics, natural products with pleiotropic effects that simultaneously target numerous signaling pathways appear to be ideal substitutes for cancer therapy. With this in mind, we take a look at the current status, impact and potential of known compounds as golden phytochemicals on key signaling pathways in tumors, focusing on the Notch pathway. This review may be useful for discovering new molecular targets for safe and efficient cancer therapy with natural chemotherapeutics.
Collapse
Affiliation(s)
| | - Nastaran Hashemzadeh
- Pharmaceutical Analysis Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajjad Alimohammadvand
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Sharifi-Azad
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, US
| | - Rana Jahanban-Esfahlan
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Sun Y, Tong H, Chu X, Li Y, Zhang J, Ding Y, Zhang S, Gui X, Chen C, Xu M, Li Z, Gardiner EE, Andrews RK, Zeng L, Xu K, Qiao J. Notch1 regulates hepatic thrombopoietin production. Blood 2024; 143:2778-2790. [PMID: 38603632 DOI: 10.1182/blood.2023023559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024] Open
Abstract
ABSTRACT Notch signaling regulates cell-fate decisions in several developmental processes and cell functions. However, the role of Notch in hepatic thrombopoietin (TPO) production remains unclear. We noted thrombocytopenia in mice with hepatic Notch1 deficiency and so investigated TPO production and other features of platelets in these mice. We found that the liver ultrastructure and hepatocyte function were comparable between control and Notch1-deficient mice. However, the Notch1-deficient mice had significantly lower plasma TPO and hepatic TPO messenger RNA levels, concomitant with lower numbers of platelets and impaired megakaryocyte differentiation and maturation, which were rescued by addition of exogenous TPO. Additionally, JAK2/STAT3 phosphorylation was significantly inhibited in Notch1-deficient hepatocytes, consistent with the RNA-sequencing analysis. JAK2/STAT3 phosphorylation and TPO production was also impaired in cultured Notch1-deficient hepatocytes after treatment with desialylated platelets. Consistently, hepatocyte-specific Notch1 deletion inhibited JAK2/STAT3 phosphorylation and hepatic TPO production induced by administration of desialylated platelets in vivo. Interestingly, Notch1 deficiency downregulated the expression of HES5 but not HES1. Moreover, desialylated platelets promoted the binding of HES5 to JAK2/STAT3, leading to JAK2/STAT3 phosphorylation and pathway activation in hepatocytes. Hepatocyte Ashwell-Morell receptor (AMR), a heterodimer of asialoglycoprotein receptor 1 [ASGR1] and ASGR2, physically associates with Notch1, and inhibition of AMR impaired Notch1 signaling activation and hepatic TPO production. Furthermore, blockage of Delta-like 4 on desialylated platelets inhibited hepatocyte Notch1 activation and HES5 expression, JAK2/STAT3 phosphorylation, and subsequent TPO production. In conclusion, our study identifies a novel regulatory role of Notch1 in hepatic TPO production, indicating that it might be a target for modulating TPO level.
Collapse
Affiliation(s)
- Yueyue Sun
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Huan Tong
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Xiang Chu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Yingying Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Jie Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Yangyang Ding
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Sixuan Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Xiang Gui
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Chong Chen
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Mengdi Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Elizabeth E Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Robert K Andrews
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| |
Collapse
|
4
|
Xie D, Ouyang S. The role and mechanisms of macrophage polarization and hepatocyte pyroptosis in acute liver failure. Front Immunol 2023; 14:1279264. [PMID: 37954583 PMCID: PMC10639160 DOI: 10.3389/fimmu.2023.1279264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Acute liver failure (ALF) is a severe liver disease caused by disruptions in the body's immune microenvironment. In the early stages of ALF, Kupffer cells (KCs) become depleted and recruit monocytes derived from the bone marrow or abdomen to replace the depleted macrophages entering the liver. These monocytes differentiate into mature macrophages, which are activated in the immune microenvironment of the liver and polarized to perform various functions. Macrophage polarization can occur in two directions: pro-inflammatory M1 macrophages and anti-inflammatory M2 macrophages. Controlling the ratio and direction of M1 and M2 in ALF can help reduce liver injury. However, the liver damage caused by pyroptosis should not be underestimated, as it is a caspase-dependent form of cell death. Inhibiting pyroptosis has been shown to effectively reduce liver damage induced by ALF. Furthermore, macrophage polarization and pyroptosis share common binding sites, signaling pathways, and outcomes. In the review, we describe the role of macrophage polarization and pyroptosis in the pathogenesis of ALF. Additionally, we preliminarily explore the relationship between macrophage polarization and pyroptosis, as well as their effects on ALF.
Collapse
Affiliation(s)
| | - Shi Ouyang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Department of Infectious Diseases, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Jo HR, Hwang J, Jeong JH. MicroRNA miR-214-5p induces senescence of microvascular endothelial cells by targeting the JAG1/Notch signaling pathway. Noncoding RNA Res 2023; 8:385-391. [PMID: 37260583 PMCID: PMC10227379 DOI: 10.1016/j.ncrna.2023.05.002] [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: 02/10/2023] [Revised: 04/10/2023] [Accepted: 05/01/2023] [Indexed: 06/02/2023] Open
Abstract
During cellular senescence, irreversible cell cycle arrest is accompanied by morphological and genetic alterations. MicroRNAs (miRNAs) play a critical role in regulating senescence by modulating the abundance of crucial senescence regulatory proteins. Therefore, to identify novel senescence-associated miRNAs, we analyzed differentially expressed miRNAs in microvascular endothelial cells (MVEC). Among the 80 differentially expressed miRNAs in replicative senescent MVECs, 16 miRNAs of unknown gene ontology were used in the senescence-associated β-galactosidase assay. Thus, we identified miR-214-5p as having high senescence-inducing activity, inhibiting the proliferation and angiogenesis activity of MVECs. To reveal the senescence-regulating mechanism of miR-214-5p, we searched for target genes through sequence- and literature-based analysis. Molecular manipulation of miR-214-5p demonstrated that miR-214-5p regulated the expression and function of Jagged 1 (JAG1) in senescent MVECs. Silencing JAG1 or downstream genes of JAG1-Notch signaling, accelerated the senescence of MVECs. Additionally, ectopic overexpression of JAG1 reversed the senescence-inducing activity of miR-214-5p. In conclusion, we identified miR-214-5p as a senescence-associated miRNA. Targeting miR-214-5p may be a potential strategy to delay vascular aging and overcome the detrimental effects of senescence and age-related diseases.
Collapse
Affiliation(s)
- Hye-ram Jo
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Science, Seoul, 01812, South Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, 34113, South Korea
| | - Jiwon Hwang
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Science, Seoul, 01812, South Korea
- Department of Life Sciences, Korea University, Seoul, 02841, South Korea
| | - Jae-Hoon Jeong
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Science, Seoul, 01812, South Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, 34113, South Korea
| |
Collapse
|
6
|
Rothzerg E, Erber WN, Gibbons CLMH, Wood D, Xu J. Osteohematology: To be or Notch to be. J Cell Physiol 2023. [PMID: 37269472 DOI: 10.1002/jcp.31042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/08/2023] [Accepted: 05/06/2023] [Indexed: 06/05/2023]
Abstract
Osteohematology is an emerging research field that studies the crosstalk between hematopoietic and bone stromal cells, to elucidate the mechanisms of hematological and skeletal malignancies and diseases. The Notch is an evolutionary conserved developmental signaling pathway, with critical roles in embryonic development by controlling cell proliferation and differentiation. However, the Notch pathway is also critically involved in cancer initiation and progression, such as osteosarcoma, leukemia, and multiple myeloma. The Notch-mediated malignant cells dysregulate bone and bone marrow cells in the tumour microenvironment, resulting in disorders ranging from osteoporosis to bone marrow dysfunction. To date, the complex interplay of Notch signaling molecules in hematopoietic and bone stromal cells is still poorly understood. In this mini-review, we summarize the crosstalk between cells in bone and bone marrow and their influence under the Notch signaling pathway in physiological conditions and in tumour microenvironment.
Collapse
Affiliation(s)
- Emel Rothzerg
- School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Wendy N Erber
- School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
- PathWest Laboratory Medicine, Nedlands, Western Australia, Australia
| | - Christopher L M H Gibbons
- Orthopaedics Oncology, Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Trust, Oxford, UK
| | - David Wood
- Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
7
|
Xiao X, Zhang Q. Asiaticoside conveys an antifibrotic effect by inhibiting activation of hepatic stellate cells via the Jagged-1/Notch-1 pathway. J Nat Med 2023; 77:128-136. [PMID: 36169781 DOI: 10.1007/s11418-022-01653-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/15/2022] [Indexed: 01/06/2023]
Abstract
The aim of this study was to investigate the underlying protective mechanisms of asiaticoside (AS) against liver fibrosis (LF) both in vivo and in vitro. A rat model with carbon tetrachloride (CCl4)-induced liver fibrosis is employed to verify the effect and mechanism of AS on the process of liver fibrosis in vivo experiment. Hematoxylin/eosin and sirius red staining was conducted to assess the severity of liver injury and fibrosis. Further, the serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin (ALB), glutamyl transferase (GGT), and total bilirubin (TBil) were measured. In addition, LX2 cells were cultured for vitro experiment to investigate the influence of AS on hepatic stellate cells (HSCs). Overproduction of α-smooth muscle actin and type I collagen is characteristic of LF and HSCs, as determined by immunohistochemical and Western blot analyses. The expression levels of molecules associated with the Notch signaling pathway (i.e., Notch-1, Jagged-1, and Delta-like-4) were assessed by Western blot analysis. The results revealed that AS attenuated LF, as defined by reduced deposition of collagen, expression of α-smooth muscle actin and collagen type 1, and expression of biochemical parameters (alanine aminotransferase, aspartate aminotransferase, and hydroxyproline). Notably, AS suppressed the expression levels of Notch-1, Jagged-1, and Delta-like-4 in activated HSCs and LF. Collectively, these results demonstrate that AS prevented the progression of LF by modulating the Notch signaling pathway, indicating that AS has potential therapeutic effects against LF.
Collapse
Affiliation(s)
- Xianhong Xiao
- Department of Infectious Disease, The People's Hospital of Yuhuan, The Yuhuan Branch of the First Affiliated Hospital With Wenzhou Medical University, 18 Changle Road, Yucheng Street, Yuhuan, 317600, Zhejiang, China.
| | - Qiang Zhang
- Department of Infectious Disease, The People's Hospital of Yuhuan, The Yuhuan Branch of the First Affiliated Hospital With Wenzhou Medical University, 18 Changle Road, Yucheng Street, Yuhuan, 317600, Zhejiang, China
| |
Collapse
|
8
|
Wu X, He W, Mu X, Liu Y, Deng J, Liu Y, Nie X. Macrophage polarization in diabetic wound healing. BURNS & TRAUMA 2022; 10:tkac051. [PMID: 36601058 PMCID: PMC9797953 DOI: 10.1093/burnst/tkac051] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/22/2022] [Indexed: 12/31/2022]
Abstract
Impaired wound healing is one of the severe complications of diabetes. Macrophages have been shown to play a vital role in wound healing. In different wound environments, macrophages are classified into two phenotypes: classically activated macrophages and alternatively activated macrophages. Dysregulation of macrophage phenotypes leads to severely impaired wound healing in diabetes. Particularly, uncontrolled inflammation and abnormal macrophage phenotype are important reasons hindering the closure of diabetic wounds. This article reviews the functions of macrophages at various stages of wound healing, the relationship between macrophage phenotypic dysregulation and diabetic wound healing and the mechanism of macrophage polarization in diabetic wound healing. New therapeutic drugs targeting phagocyte polarization to promote the healing of diabetic wounds might provide a new strategy for treating chronic diabetic wound healing.
Collapse
Affiliation(s)
- Xingqian Wu
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Wenjie He
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Xingrui Mu
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Ye Liu
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Junyu Deng
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Yiqiu Liu
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China
- Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, Zunyi Medical University, Zunyi 563000, China
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563000, China
- Cancer and Ageing Research Program, School of Biomedical Sciences, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane 4102, Australia
| |
Collapse
|
9
|
Giuli MV, Mancusi A, Giuliani E, Screpanti I, Checquolo S. Notch signaling in female cancers: a multifaceted node to overcome drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 4:805-836. [PMID: 35582386 PMCID: PMC8992449 DOI: 10.20517/cdr.2021.53] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/24/2022]
Abstract
Drug resistance is one of the main challenges in cancer therapy, including in the treatment of female-specific malignancies, which account for more than 60% of cancer cases among women. Therefore, elucidating the underlying molecular mechanisms is an urgent need in gynecological cancers to foster novel therapeutic approaches. Notably, Notch signaling, including either receptors or ligands, has emerged as a promising candidate given its multifaceted role in almost all of the hallmarks of cancer. Concerning the connection between Notch pathway and drug resistance in the afore-mentioned tumor contexts, several studies focused on the Notch-dependent regulation of the cancer stem cell (CSC) subpopulation or the induction of the epithelial-to-mesenchymal transition (EMT), both features implicated in either intrinsic or acquired resistance. Indeed, the present review provides an up-to-date overview of the published results on Notch signaling and EMT- or CSC-driven drug resistance. Moreover, other drug resistance-related mechanisms are examined such as the involvement of the Notch pathway in drug efflux and tumor microenvironment. Collectively, there is a long way to go before every facet will be fully understood; nevertheless, some small pieces are falling neatly into place. Overall, the main aim of this review is to provide strong evidence in support of Notch signaling inhibition as an effective strategy to evade or reverse resistance in female-specific cancers.
Collapse
Affiliation(s)
- Maria V Giuli
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome 00161, Italy
| | - Angelica Mancusi
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome 00161, Italy
| | - Eugenia Giuliani
- Scientific Direction, San Gallicano Dermatological Institute IRCCS, Rome 00144, Italy
| | - Isabella Screpanti
- Laboratory of Molecular Pathology, Department of Molecular Medicine, Sapienza University, Rome 00161, Italy
| | - Saula Checquolo
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University, Latina 04100, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy
| |
Collapse
|
10
|
Bian W, Tang M, Jiang H, Xu W, Hao W, Sui Y, Hou Y, Nie L, Zhang H, Wang C, Li N, Wang J, Qin J, Wu L, Ma X, Chen J, Wang W, Li X. Low-density-lipoprotein-receptor-related protein 1 mediates Notch pathway activation. Dev Cell 2021; 56:2902-2919.e8. [PMID: 34626540 DOI: 10.1016/j.devcel.2021.09.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 08/18/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022]
Abstract
The Notch signaling pathway controls cell growth, differentiation, and fate decisions, and its dysregulation has been linked to various human genetic disorders and cancers. To comprehensively understand the global organization of the Notch pathway and identify potential drug targets for Notch-related diseases, we established a protein interaction landscape for the human Notch pathway. By combining and analyzing genetic and phenotypic data with bioinformatics analysis, we greatly expanded this pathway and identified many key regulators, including low-density-lipoprotein-receptor-related protein 1 (LRP1). We demonstrated that LRP1 mediates the ubiquitination chain linkage switching of Delta ligands, which further affects ligand recycling, membrane localization, and stability. LRP1 inhibition led to Notch signaling inhibition and decreased tumorigenesis in leukemia models. Our study provides a glimpse into the Notch pathway interaction network and uncovers LRP1 as one critical regulator of the Notch pathway, as well as a possible therapeutic target for Notch-related cancers.
Collapse
Affiliation(s)
- Weixiang Bian
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Mengfan Tang
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Hua Jiang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Wenyan Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Wanyu Hao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Science, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Yue Sui
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yingnan Hou
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Litong Nie
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Huimin Zhang
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Chao Wang
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Nan Li
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Jiadong Wang
- Institute of Systems Biomedicine, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, People's Republic of China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Lianfeng Wu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Science, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Xianjue Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| | - Junjie Chen
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA.
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA.
| | - Xu Li
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China.
| |
Collapse
|
11
|
Chen W, Liu Y, Chen J, Ma Y, Song Y, Cen Y, You M, Yang G. The Notch signaling pathway regulates macrophage polarization in liver diseases. Int Immunopharmacol 2021; 99:107938. [PMID: 34371331 DOI: 10.1016/j.intimp.2021.107938] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/16/2022]
Abstract
The liver is not only the main metabolic site of exogenous compounds and drugs, but also an important immune organ in the human body. When a large number of nonself substances (such as drugs, alcohol, pathogens, microorganisms and their metabolites) enter the liver, they will cause serious liver diseases, including liver fibrosis, liver cirrhosis, liver failure, and hepatocellular carcinoma (HCC). Macrophages are the first line of defense against the invasion of exogenous pathogens and significant cellular components of the innate immune system. Macrophages have strong heterogeneity and plasticity. When different pathogens invade the body, they cause different types of polarization of macrophages through different molecular mechanisms. Notch signaling is considered to be the key regulator of the biological function of macrophages. Activating Notch signaling can regulate the differentiation of macrophages into M1 and play a role in promoting inflammation and antitumor activity, while blocking Notch signaling can polarize macrophages to M2, suppressing inflammation and promoting tumor growth. However, there are few studies on regulation of macrophage polarization by the Notch signaling pathway in liver diseases. Therefore, in this review, we will introduce the role of the Notch signaling pathway in regulating macrophage polarization in liver diseases.
Collapse
Affiliation(s)
- Wenyan Chen
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yining Liu
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Jing Chen
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yemei Ma
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yawen Song
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yanli Cen
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Mingdan You
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Guanghong Yang
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou 550004, China.
| |
Collapse
|
12
|
He H, Shao X, Li Y, Gihu R, Xie H, Zhou J, Yan H. Targeting Signaling Pathway Networks in Several Malignant Tumors: Progresses and Challenges. Front Pharmacol 2021; 12:675675. [PMID: 34135756 PMCID: PMC8203325 DOI: 10.3389/fphar.2021.675675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/18/2021] [Indexed: 12/22/2022] Open
Abstract
Malignant tumors remain the health problem of highest concern among people worldwide due to its high mortality and recurrence. Lung, gastric, liver, colon, and breast cancers are among the top five malignant tumors in terms of morbidity and mortality. In cancer biology, aberrant signaling pathway regulation is a prevalent theme that drives the generation, metastasis, invasion, and other processes of all malignant tumors. The Wnt/β-catenin, PI3K/AKT/mTOR, Notch and NF-kB pathways are widely concerned and signal crosstalks exist in the five solid tumors. This review provides an innovative summary of the recent progress in research on these signaling pathways, the underlying mechanism of the molecules involved in these pathways, and the important role of some miRNAs in tumor-related signaling pathways. It also presents a brief review of the antitumor molecular drugs that target these signaling pathways. This review may provide a theoretical basis for the study of the molecular biological mechanism of malignant tumors and vital information for the development of new treatment strategies with a focus on efficacy and the reduction of side effects.
Collapse
Affiliation(s)
- Hongdan He
- Qinghai Tibet Plateau Research Institute, Southwest Minzu University, Chengdu, China
| | - Xiaoni Shao
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Yanan Li
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Ribu Gihu
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Haochen Xie
- Qinghai Tibet Plateau Research Institute, Southwest Minzu University, Chengdu, China
| | - Junfu Zhou
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| |
Collapse
|
13
|
Guo S, Quan S, Zou S. Roles of the Notch Signaling Pathway in Ovarian Functioning. Reprod Sci 2021; 28:2770-2778. [PMID: 34008156 DOI: 10.1007/s43032-021-00610-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/05/2021] [Indexed: 11/30/2022]
Abstract
The Notch signaling pathway regulates cell invasion, adhesion, proliferation, apoptosis, and differentiation via cell-to-cell interactions and plays important physiological roles in the ovary. This review summarizes current knowledge about the Notch signaling pathway in relation to ovarian functions and reveals the potential underlying mechanisms. We conducted an in-depth review of relevant literature to determine the current status of research into the Notch signaling pathway in relation to ovarian functioning and reveal potential underlying mechanisms. The activation of different Notch receptors promotes the formation of primordial follicles and proliferation of granulosa cells and inhibits steroid secretion. Abnormal regulation of the Notch signaling pathway or direct mutations might lead to over-activation or under-activation of the receptors, resulting in Notch upregulation or downregulation. It can also disrupt the normal physiological functions of the ovary. The lncRNA HOTAIR and growth hormones improved premature ovarian failure (POF) and promoted follicle maturation in a mouse model of POF by upregulating Notch1 expression. They also stimulated the Notch1 signaling pathway, increased the level of plasma estradiol, and decreased the level of plasma follicle-stimulating hormone. Thus, Notch1 could serve as a novel therapeutic target for POF. Several studies have reported multiple roles of Notch in regulating female primordial follicle formation and follicle maturation. Direct mutations in Notch-related molecules or abnormal gene regulation in the signaling pathway can lead to ovarian dysfunction. However, the underlying mechanisms are not fully understood.
Collapse
Affiliation(s)
- Shuhan Guo
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Song Quan
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - Siyi Zou
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Large-scale circular RNA deregulation in T-ALL: unlocking unique ectopic expression of molecular subtypes. Blood Adv 2021; 4:5902-5914. [PMID: 33259601 DOI: 10.1182/bloodadvances.2020002337] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/20/2020] [Indexed: 12/25/2022] Open
Abstract
Circular RNAs (circRNAs) are stable RNA molecules that can drive cancer through interactions with microRNAs and proteins and by the expression of circRNA encoded peptides. The aim of the study was to define the circRNA landscape and potential impact in T-cell acute lymphoblastic leukemia (T-ALL). Analysis by CirComPara of RNA-sequencing data from 25 T-ALL patients, immature, HOXA overexpressing, TLX1, TLX3, TAL1, or LMO2 rearranged, and from thymocyte populations of human healthy donors disclosed 68 554 circRNAs. Study of the top 3447 highly expressed circRNAs identified 944 circRNAs with significant differential expression between malignant T cells and normal counterparts, with most circRNAs displaying increased expression in T-ALL. Next, we defined subtype-specific circRNA signatures in molecular genetic subgroups of human T-ALL. In particular, circZNF609, circPSEN1, circKPNA5, and circCEP70 were upregulated in immature, circTASP1, circZBTB44, and circBACH1 in TLX3, circHACD1, and circSTAM in HOXA, circCAMSAP1 in TLX1, and circCASC15 in TAL-LMO. Backsplice sequences of 14 circRNAs ectopically expressed in T-ALL were confirmed, and overexpression of circRNAs in T-ALL with specific oncogenic lesions was substantiated by quantification in a panel of 13 human cell lines. An oncogenic role of circZNF609 in T-ALL was indicated by decreased cell viability upon silencing in vitro. Furthermore, functional predictions identified circRNA-microRNA gene axes informing modes of circRNA impact in molecular subtypes of human T-ALL.
Collapse
|
15
|
Gratton R, Tricarico PM, d’Adamo AP, Bianco AM, Moura R, Agrelli A, Brandão L, Zupin L, Crovella S. Notch Signaling Regulation in Autoinflammatory Diseases. Int J Mol Sci 2020; 21:E8847. [PMID: 33238371 PMCID: PMC7700323 DOI: 10.3390/ijms21228847] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/13/2020] [Accepted: 11/21/2020] [Indexed: 12/22/2022] Open
Abstract
Notch pathway is a highly conserved intracellular signaling route that modulates a vast variety of cellular processes including proliferation, differentiation, migration, cell fate and death. Recently, the presence of a strict crosstalk between Notch signaling and inflammation has been described, although the precise molecular mechanisms underlying this interplay have not yet been fully unravelled. Disruptions in Notch cascade, due both to direct mutations and/or to an altered regulation in the core components of Notch signaling, might lead to hypo- or hyperactivation of Notch target genes and signaling molecules, ultimately contributing to the onset of autoinflammatory diseases. To date, alterations in Notch signaling have been reported as associated with three autoinflammatory disorders, therefore, suggesting a possible role of Notch in the pathogenesis of the following diseases: hidradenitis suppurativa (HS), Behçet disease (BD), and giant cell arteritis (GCA). In this review, we aim at better characterizing the interplay between Notch and autoinflammatory diseases, trying to identify the role of this signaling route in the context of these disorders.
Collapse
Affiliation(s)
- Rossella Gratton
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Paola Maura Tricarico
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Adamo Pio d’Adamo
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
- Department of Medical Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Anna Monica Bianco
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Ronald Moura
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Almerinda Agrelli
- Department of Pathology, Federal University of Pernambuco, Recife 50670-901, Brazil;
| | - Lucas Brandão
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Luisa Zupin
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Sergio Crovella
- Department of Biological and Environmental Sciences, College of Arts and Sciences, University of Qatar, Doha 2713, Qatar;
| |
Collapse
|
16
|
Gratton R, Tricarico PM, Moltrasio C, Lima Estevão de Oliveira AS, Brandão L, Marzano AV, Zupin L, Crovella S. Pleiotropic Role of Notch Signaling in Human Skin Diseases. Int J Mol Sci 2020; 21:E4214. [PMID: 32545758 PMCID: PMC7353046 DOI: 10.3390/ijms21124214] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Notch signaling orchestrates the regulation of cell proliferation, differentiation, migration and apoptosis of epidermal cells by strictly interacting with other cellular pathways. Any disruption of Notch signaling, either due to direct mutations or to an aberrant regulation of genes involved in the signaling route, might lead to both hyper- or hypo-activation of Notch signaling molecules and of target genes, ultimately inducing the onset of skin diseases. The mechanisms through which Notch contributes to the pathogenesis of skin diseases are multiple and still not fully understood. So far, Notch signaling alterations have been reported for five human skin diseases, suggesting the involvement of Notch in their pathogenesis: Hidradenitis Suppurativa, Dowling Degos Disease, Adams-Oliver Syndrome, Psoriasis and Atopic Dermatitis. In this review, we aim at describing the role of Notch signaling in the skin, particularly focusing on the principal consequences associated with its alterations in these five human skin diseases, in order to reorganize the current knowledge and to identify potential cellular mechanisms in common between these pathologies.
Collapse
Affiliation(s)
- Rossella Gratton
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (L.Z.); (S.C.)
- Department of Medical Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Paola Maura Tricarico
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (L.Z.); (S.C.)
| | - Chiara Moltrasio
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (C.M.); (A.V.M.)
| | | | - Lucas Brandão
- Department of Pathology, Federal University of Pernambuco, Recife 50670-901, Brazil;
| | - Angelo Valerio Marzano
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (C.M.); (A.V.M.)
| | - Luisa Zupin
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (L.Z.); (S.C.)
| | - Sergio Crovella
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (L.Z.); (S.C.)
- Department of Medical Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| |
Collapse
|
17
|
Bill M, Pathmanathan A, Karunasiri M, Shen C, Burke MH, Ranganathan P, Papaioannou D, Zitzer NC, Snyder K, LaRocco A, Walker AE, Brannan ZJ, Nalin AP, Freud AG, Dikov MM, Zhang X, Bloomfield CD, Garzon R, Dorrance AM. EGFL7 Antagonizes NOTCH Signaling and Represents a Novel Therapeutic Target in Acute Myeloid Leukemia. Clin Cancer Res 2020; 26:669-678. [PMID: 31672772 DOI: 10.1158/1078-0432.ccr-19-2479] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/24/2019] [Accepted: 10/21/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE EGF-like domain 7 (EGFL7) is a secreted protein and recently has been shown to play an important role in acute myeloid leukemia (AML); however, the underlying mechanism by which EGFL7 promotes leukemogenesis is largely unknown. EXPERIMENTAL DESIGN Using an antibody interaction array, we measured the ability of EGFL7 to bind directly approximately 400 proteins expressed by primary AML blasts. Primary patient samples were stimulated in vitro with recombinant EGFL7 (rEGFL7) or anti-EGFL7 blocking antibody to assess alterations in downstream signaling and the ability to effect blast differentiation and survival. We treated three independent AML models with anti-EGFL7 or IgG1 control to determine whether anti-EGFL7 could prolong survival in vivo. RESULTS We found EGFL7 significantly binds several signaling proteins important for normal and malignant hematopoiesis including NOTCH. Stimulation of AML blasts with rEGFL7 reduced NOTCH intracellular domain and NOTCH target gene expression while treatment with an anti-EGFL7 blocking antibody resulted in reactivation of NOTCH signaling, increased differentiation, and apoptosis. Competitive ligand-binding assays showed rEGFL7 inhibits DELTA-like (DLL) 4-mediated NOTCH activation while anti-EGFL7 combined with DLL4 significantly increased NOTCH activation and induced apoptosis. Using three different AML mouse models, we demonstrated that in vivo treatment with anti-EGFL7 alone results in increased survival. CONCLUSIONS Our data demonstrate that EGFL7 contributes to NOTCH silencing in AML by antagonizing canonical NOTCH ligand binding. Reactivation of NOTCH signaling in vivo using anti-EGFL7 results in prolonged survival of leukemic mice, supporting the use of EGFL7 as a novel therapeutic target in AML.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/pharmacology
- Apoptosis
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Cell Differentiation
- Cell Line, Tumor
- Cell Proliferation
- Disease Models, Animal
- EGF Family of Proteins/genetics
- EGF Family of Proteins/metabolism
- Female
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Receptors, Notch/antagonists & inhibitors
- Receptors, Notch/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Marius Bill
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Aparna Pathmanathan
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Malith Karunasiri
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Changxian Shen
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Matthew H Burke
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Parvathi Ranganathan
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Dimitrios Papaioannou
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Nina C Zitzer
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Katiri Snyder
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Allison LaRocco
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Allison E Walker
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Zachary J Brannan
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Ansel P Nalin
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Aharon G Freud
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Mikhail M Dikov
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Xiaoli Zhang
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Clara D Bloomfield
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Ramiro Garzon
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Adrienne M Dorrance
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.
- Division of Hematology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
18
|
Zhu Q, Hu L, Guo Y, Xiao Z, Xu Q, Tong X. FBW7 in hematological tumors. Oncol Lett 2020; 19:1657-1664. [PMID: 32194657 PMCID: PMC7039162 DOI: 10.3892/ol.2020.11264] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
Abstract
F-box and WD repeat domain-containing protein 7 (FBW7), also known as FBXW7, AGO or hCDC4, is an F-box protein with seven tandem WD40 repeats. FBW7 is a key substrate recognition subunit of the Skp1-Cul1-F-box-protein E3 ubiquitin ligase. FBW7 targets for ubiquitination and destruction of numerous crucial transcription factors and protooncogenes, including cyclin E, c-Myc, c-Jun, Notch and MCL-1. FBW7 is a well-characterized tumor suppressor, and its gene is frequently mutated or deleted in various types of human cancer, including colorectal cancer, gastric cancer, ovarian cancer and different types of leukemia. Accumulating evidence indicates that the aberrant expression of FBW7 is involved in the development of hematological tumors, including T cell acute lymphoblastic leukemia, adult T cell leukemia/lymphoma, chronic lymphocytic leukemia and multiple myeloma. The present review will describe the latest findings on the role of FBW7 in hematological tumors, in order to identify a novel target for future therapies.
Collapse
Affiliation(s)
- Qiaojuan Zhu
- The Second Clinical Medical Department, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, P.R. China
| | - Linjun Hu
- Medical Department, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Yang Guo
- Graduate Department, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Zunqiang Xiao
- The Second Clinical Medical Department, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, P.R. China
| | - Qiuran Xu
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang 310014, P.R. China
| | - Xiangmin Tong
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
19
|
Gu G, Gao T, Zhang L, Chen X, Pang Q, Wang Y, Wang D, Li J, Liu Q. NKAP alters tumor immune microenvironment and promotes glioma growth via Notch1 signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:291. [PMID: 31277684 PMCID: PMC6612223 DOI: 10.1186/s13046-019-1281-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
Abstract
Background Glioma is one of the most aggressive malignant brain tumors which is characterized with highly infiltrative growth and poor prognosis. NKAP (NF-κB activating protein) is a widely expressed 415-amino acid nuclear protein that is overexpressed by gliomas, but its function in glioma was still unknown. Methods CCK8 and EDU assay was used to examine the cell viability in vitro, and the xenograft models in nude mice were established to explore the roles of NAKP in vivo. The expressions of NKAP, Notch1 and SDF-1 were analyzed by immunofluorescence analysis. The expression of NKAP and Notch1 in glioma and normal human brain samples were analyzed by immunohistochemical analysis. In addition, CHIP, Gene chip, western blot, flow cytometry, immunofluorescence, ELISA and luciferase assay were used to investigate the internal connection between NKAP and Notch1. Results Here we showed that overexpression of NKAP in gliomas could promote tumor growth by contributing to a Notch1-dependent immune-suppressive tumor microenvironment. Downregulation of NKAP in gliomas had abrogated tumor growth and invasion in vitro and in vivo. Interestingly, compared to the control group, inhibiting NKAP set up obstacles to tumor-associated macrophage (TAM) polarization and recruitment by decreasing the secretion of SDF-1 and M-CSF. To identify the potential mechanisms involved, we performed RNA sequencing analysis and found that Notch1 appeared to positively correlate with the expression of NKAP. Furthermore, we proved that NKAP performed its function via directly binding to Notch1 promoter and trans-activating it. Notch1 inhibition could alleviate NKAP’s gliomagenesis effects. Conclusion these observations suggest that NKAP promotes glioma growth by TAM chemoattraction through upregulation of Notch1 and this finding introduces the potential utility of NKAP inhibitors for glioma therapy. Electronic supplementary material The online version of this article (10.1186/s13046-019-1281-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guangyan Gu
- Department of Histology and Embryology, School of Basic Medical Science, Shandong University Cheeloo College of Medicine, 44# Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.,Assisted Reproductive Centre, Shandong Maternity and Child Health Care Hospital, Jinan, Shandong, China
| | - Taihong Gao
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Lu Zhang
- Department of Peripheral Vascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiuyang Chen
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Qi Pang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Yanan Wang
- Department of Histology and Embryology, School of Basic Medical Science, Shandong University Cheeloo College of Medicine, 44# Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Dan Wang
- Department of Histology and Embryology, School of Basic Medical Science, Shandong University Cheeloo College of Medicine, 44# Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Jie Li
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Qian Liu
- Department of Histology and Embryology, School of Basic Medical Science, Shandong University Cheeloo College of Medicine, 44# Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Chronic lymphocytic leukemia is heterogeneous disease characterized by a variable clinical course that is greatly influenced by various patient and disease characteristics. Over the last two decades, advent of new diagnostic methodologies has led to the identification of several factors of prognostic and predictive relevance. Furthermore, recent advances in next-generation sequencing techniques has identified recurrent novel mutations in NOTCH1, SF3B1, BIRC3, and ATM genes whose role as prognostic and predictive markers is currently being investigated. These biologic markers carry new prognostic information and their incorporation into prognostic scoring systems will likely lead to refined multi-parameter risk models. RECENT FINDINGS While the prognostic impact of many of the most commonly used markers on clinical outcomes in patients treated with chemo-immunotherapy is well documented, it is important to review their predictive and prognostic role in the era of novel targeted therapies. This article will discuss the currently available information on the clinical relevance of prognostic markers in patients treated with novel targeted therapies.
Collapse
Affiliation(s)
- Prajwal Boddu
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 428, Houston, TX, 77030, USA
| | - Alessandra Ferrajoli
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 428, Houston, TX, 77030, USA.
| |
Collapse
|
21
|
Zebrafish in Translational Cancer Research: Insight into Leukemia, Melanoma, Glioma and Endocrine Tumor Biology. Genes (Basel) 2017; 8:genes8090236. [PMID: 28930163 PMCID: PMC5615369 DOI: 10.3390/genes8090236] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/08/2017] [Accepted: 09/14/2017] [Indexed: 02/06/2023] Open
Abstract
Over the past 15 years, zebrafish have emerged as a powerful tool for studying human cancers. Transgenic techniques have been employed to model different types of tumors, including leukemia, melanoma, glioblastoma and endocrine tumors. These models present histopathological and molecular conservation with their human cancer counterparts and have been fundamental for understanding mechanisms of tumor initiation and progression. Moreover, xenotransplantation of human cancer cells in embryos or adult zebrafish offers the advantage of studying the behavior of human cancer cells in a live organism. Chemical-genetic screens using zebrafish embryos have uncovered novel druggable pathways and new therapeutic strategies, some of which are now tested in clinical trials. In this review, we will report on recent advances in using zebrafish as a model in cancer studies—with specific focus on four cancer types—where zebrafish has contributed to novel discoveries or approaches to novel therapies.
Collapse
|
22
|
Chandrakesan P, Yao J, Qu D, May R, Weygant N, Ge Y, Ali N, Sureban SM, Gude M, Vega K, Bannerman-Menson E, Xia L, Bronze M, An G, Houchen CW. Dclk1, a tumor stem cell marker, regulates pro-survival signaling and self-renewal of intestinal tumor cells. Mol Cancer 2017; 16:30. [PMID: 28148261 PMCID: PMC5286867 DOI: 10.1186/s12943-017-0594-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/18/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND More than 80% of intestinal neoplasia is associated with the adenomatous polyposis coli (APC) mutation. Doublecortin-like kinase 1 (Dclk1), a kinase protein, is overexpressed in colorectal cancer and specifically marks tumor stem cells (TSCs) that self-renew and increased the tumor progeny in Apc Min/+ mice. However, the role of Dclk1 expression and its contribution to regulating pro-survival signaling for tumor progression in Apc mutant cancer is poorly understood. METHODS We analyzed DCLK1 and pro-survival signaling gene expression datasets of 329 specimens from TCGA Colon Adenocarcinoma Cancer Data. The network of DCLK1 and pro-survival signaling was analyzed utilizing the GeneMANIA database. We examined the expression levels of Dclk1 and other stem cell-associated markers, pro-survival signaling pathways, cell self-renewal in the isolated intestinal epithelial cells of Apc Min/+ mice with high-grade dysplasia and adenocarcinoma. To determine the functional role of Dclk1 for tumor progression, we knocked down Dclk1 and determined the pro-survival signaling pathways and stemness. We used siRNA technology to gene silence pro-survival signaling in colon cancer cells in vitro. We utilized FACS, IHC, western blot, RT-PCR, and clonogenic (self-renewal) assays. RESULTS We found a correlation between DCLK1 and pro-survival signaling expression. The expression of Dclk1 and stem cell-associated markers Lgr5, Bmi1, and Musashi1 were significantly higher in the intestinal epithelial cells of Apc Min/+ mice than in wild-type controls. Intestinal epithelial cells of Apc Min/+ mice showed increased expression of pro-survival signaling, pluripotency and self-renewal ability. Furthermore, the enteroids formed from the intestinal Dclk1+ cells of Apc Min/+ mice display higher pluripotency and pro-survival signaling. Dclk1 knockdown in Apc Min/+ mice attenuates intestinal adenomas and adenocarcinoma, and decreases pro-survival signaling and self-renewal. Knocking down RELA and NOTCH1 pro-survival signaling and DCLK1 in HT29 and DLD1 colon cancer cells in vitro reduced the tumor cells' ability to self-renew and survive. CONCLUSION Our results indicate that Dclk1 is essential in advancing intestinal tumorigenesis. Knocking down Dclk1 decreases tumor stemness and progression and is thus predicted to regulate pro-survival signaling and tumor cell pluripotency. This study provides a strong rationale to target Dclk1 as a treatment strategy for colorectal cancer.
Collapse
Affiliation(s)
- Parthasarathy Chandrakesan
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- OU Cancer Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK USA
| | - Jiannan Yao
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Dongfeng Qu
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- OU Cancer Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK USA
| | - Randal May
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK USA
| | - Nathaniel Weygant
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Yang Ge
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, Beijing, China
| | - Naushad Ali
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK USA
| | - Sripathi M. Sureban
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK USA
| | - Modhi Gude
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- Access Endocrine, Diabetes and Thyroid center, Oklahoma City, OK USA
| | - Kenneth Vega
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- Department of Medicine, National Jewish Health, Denver, CO USA
| | | | - Lijun Xia
- Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Michael Bronze
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Guangyu An
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, Beijing, China
| | - Courtney W. Houchen
- Division of Digestive Diseases and Nutrition, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- OU Cancer Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK USA
- COARE Biotechnology, Oklahoma City, OK USA
| |
Collapse
|
23
|
Crawford LJ, Irvine AE. The role of the CCN family of proteins in blood cancers. J Cell Commun Signal 2016; 10:197-205. [PMID: 27485291 PMCID: PMC5055499 DOI: 10.1007/s12079-016-0342-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 07/27/2016] [Indexed: 01/19/2023] Open
Abstract
Haematopoiesis is the term used to describe the production of blood cells. This is a tightly regulated hierarchical system in which mature circulating blood cells develop from a small population of haematopoietic stem (HSC) and progenitor cells within the microenvironment of the bone marrow. Molecular and genetic abnormalities arising in these stem cells lead to a block in the normal programme of proliferation and differentiation and result in the development of the blood cancers known as the leukaemias and lymphomas. Recently the regulatory role of the bone marrow microenvironment or niche has also become increasingly recognised. The interface between the bone and bone marrow (endosteum) and the region surrounding the blood vessels (perivascular) provide distinct niches harbouring quiescent HSC or proliferative HSC respectively. Current chemotherapeutic regimes can often successfully target the proliferative HSC but disease relapse occurs due to residual quiescent HSC. Understanding these developmental and regulatory processes and the associated cell communication mechanisms are thus crucial to the development of new treatment strategies. The CCN family of proteins have been recognised to play a key role in all aspects of haematopoiesis.
Collapse
Affiliation(s)
- Lisa Judith Crawford
- Haematology Research Unit, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Room OG/013, Belfast, BT9 7BL, Northern, Ireland
| | - Alexandra Elizabeth Irvine
- Haematology Research Unit, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Room OG/013, Belfast, BT9 7BL, Northern, Ireland.
| |
Collapse
|
24
|
Safdar K, Gu A, Xu X, Au V, Taylor J, Flibotte S, Moerman DG, Maine EM. UBR-5, a Conserved HECT-Type E3 Ubiquitin Ligase, Negatively Regulates Notch-Type Signaling in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2016; 6:2125-34. [PMID: 27185398 PMCID: PMC4938665 DOI: 10.1534/g3.116.027805] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/09/2016] [Indexed: 12/17/2022]
Abstract
Notch-type signaling mediates cell-cell interactions important for animal development. In humans, reduced or inappropriate Notch signaling activity is associated with various developmental defects and disease states, including cancers. Caenorhabditis elegans expresses two Notch-type receptors, GLP-1 and LIN-12. GLP-1 mediates several cell-signaling events in the embryo and promotes germline proliferation in the developing and adult gonad. LIN-12 acts redundantly with GLP-1 in certain inductive events in the embryo and mediates several cell-cell interactions during larval development. Recovery of genetic suppressors and enhancers of glp-1 or lin-12 loss- or gain-of-function mutations has identified numerous regulators of GLP-1 and LIN-12 signaling activity. Here, we report the molecular identification of sog-1, a gene identified in screens for recessive suppressors of conditional glp-1 loss-of-function mutations. The sog-1 gene encodes UBR-5, the sole C. elegans member of the UBR5/Hyd family of HECT-type E3 ubiquitin ligases. Molecular and genetic analyses indicate that the loss of ubr-5 function suppresses defects caused by reduced signaling via GLP-1 or LIN-12. In contrast, ubr-5 mutations do not suppress embryonic or larval lethality associated with mutations in a downstream transcription factor, LAG-1. In the gonad, ubr-5 acts in the receiving cells (germ cells) to limit GLP-1 signaling activity. SEL-10 is the F-box component of SCF(SEL-10) E3 ubiquitin-ligase complex that promotes turnover of Notch intracellular domain. UBR-5 acts redundantly with SEL-10 to limit Notch signaling in certain tissues. We hypothesize that UBR-5 activity limits Notch-type signaling by promoting turnover of receptor or limiting its interaction with pathway components.
Collapse
Affiliation(s)
- Komal Safdar
- Department of Biology, Syracuse University, New York 13244
| | - Anniya Gu
- Department of Biology, Syracuse University, New York 13244
| | - Xia Xu
- Department of Biology, Syracuse University, New York 13244
| | - Vinci Au
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Jon Taylor
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Stephane Flibotte
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Donald G Moerman
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | | |
Collapse
|
25
|
Amaya-Chanaga CI, Rassenti LZ. Biomarkers in chronic lymphocytic leukemia: Clinical applications and prognostic markers. Best Pract Res Clin Haematol 2016; 29:79-89. [DOI: 10.1016/j.beha.2016.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 07/07/2016] [Accepted: 08/04/2016] [Indexed: 02/01/2023]
|
26
|
Abstract
Abstract
Pediatric patients with newly diagnosed, non-Hodgkin Lymphoma (NHL) have an excellent overall survival. However, therapy regimens are associated with acute toxicity and late effects. Furthermore, patients with relapsed or refractory disease have relatively few options with proven clinical benefit. Both histologic and molecular differences exist between adult and pediatric NHL preventing simple translation of adult NHL successes into improvements in pediatric NHL treatment. This review summarizes the introduction of targeted therapies into frontline treatments for patients with anaplastic large-cell lymphoma and CD20–positive tumors, with the goal of improving overall survival while limiting both short- and long-term toxicities. In addition, newer approaches that have limited data in children but may have a significant role in how we treat pediatric NHL in the future are reviewed, which include CD19 directed therapy, Notch inhibition, the tri-functional antibody, FBTA05, and EZH2 inhibition.
Collapse
|
27
|
Abstract
Pediatric patients with newly diagnosed, non-Hodgkin Lymphoma (NHL) have an excellent overall survival. However, therapy regimens are associated with acute toxicity and late effects. Furthermore, patients with relapsed or refractory disease have relatively few options with proven clinical benefit. Both histologic and molecular differences exist between adult and pediatric NHL preventing simple translation of adult NHL successes into improvements in pediatric NHL treatment. This review summarizes the introduction of targeted therapies into frontline treatments for patients with anaplastic large-cell lymphoma and CD20–positive tumors, with the goal of improving overall survival while limiting both short- and long-term toxicities. In addition, newer approaches that have limited data in children but may have a significant role in how we treat pediatric NHL in the future are reviewed, which include CD19 directed therapy, Notch inhibition, the tri-functional antibody, FBTA05, and EZH2 inhibition.
Collapse
|
28
|
Sun R, Zhang J, Xiong M, Wei H, Tan K, Yin L, Pu Y. Altered Expression of Genes in Signaling Pathways Regulating Proliferation of Hematopoietic Stem and Progenitor Cells in Mice with Subchronic Benzene Exposure. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:9298-313. [PMID: 26262635 PMCID: PMC4555281 DOI: 10.3390/ijerph120809298] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 07/29/2015] [Accepted: 08/02/2015] [Indexed: 12/25/2022]
Abstract
Leukemias and hematopoietic disorders induced by benzene may arise from the toxicity of benzene to hematopoietic stem or progenitor cells (HS/PCs). Since there is a latency period between initial benzene exposure and the development of leukemia, subsequent impact of benzene on HS/PCs are crucial for a deeper understanding of the carcinogenicity and hematotoxicity in post-exposure stage. This study aims to explore the effects of benzene on HS/PCs and gene-expression in Wnt, Notch and Hh signaling pathways in post-exposure stage. The C3H/He mice were injected subcutaneously with benzene (0, 150, 300 mg/kg/day) for three months and were monitored for another 10 months post-exposure. The body weights were monitored, the relative organ weights, blood parameters and bone marrow smears were examined. Frequency of lineage- sca-1+ c-kit+ (LSK) cells, capability of colony forming and expression of genes in Wnt, Notch and Hedghog (Hh) signaling pathways were also analyzed. The colony formation of the progenitor cells for BFU-E, CFU-GEMM and CFU-GM was significantly decreased with increasing benzene exposure relative to controls, while no significant difference was observed in colonies for CFU-G and CFU-M. The mRNA level of cyclin D1 was increased and Notch1 and p53 were decreased in LSK cells in mice exposed to benzene but with no statistical significance. These results suggest that subsequent toxic effects of benzene on LSK cells and gene expression in Wnt, Notch and Hh signaling pathways persist in post-exposure stage and may play roles in benzene-induced hematotoxicity.
Collapse
Affiliation(s)
- Rongli Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education.
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education.
| | - Mengzhen Xiong
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education.
| | - Haiyan Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education.
| | - Kehong Tan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education.
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education.
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education
| |
Collapse
|