1
|
Tufail M, Jiang CH, Li N. Wnt signaling in cancer: from biomarkers to targeted therapies and clinical translation. Mol Cancer 2025; 24:107. [PMID: 40170063 PMCID: PMC11963613 DOI: 10.1186/s12943-025-02306-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/15/2025] [Indexed: 04/03/2025] Open
Abstract
The Wnt signaling pathway plays a crucial role in development and tissue homeostasis, regulating key cellular processes such as proliferation, differentiation, and apoptosis. However, its abnormal activation is strongly associated with tumorigenesis, metastasis, and resistance to therapy, making it a vital target for cancer treatment. This review provides a comprehensive insight into the role of Wnt signaling in cancer, examining its normal physiological functions, dysregulation in malignancies, and therapeutic potential. We emphasize the importance of predicting Wnt signaling sensitivity and identify key biomarkers across various cancer types. Additionally, we address the challenges and future prospects of Wnt-targeted therapies, including biomarker discovery, advancements in emerging technologies, and their application in clinical practice.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China.
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
2
|
Yan B, Lu Q, Gao T, Xiao K, Zong Q, Lv H, Lv G, Wang L, Liu C, Yang W, Jiang G. CD146 regulates the stemness and chemoresistance of hepatocellular carcinoma via JAG2-NOTCH signaling. Cell Death Dis 2025; 16:150. [PMID: 40032820 PMCID: PMC11876685 DOI: 10.1038/s41419-025-07470-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
CD146 plays a key role in cancer progression and metastasis. Cancer stem cells (CSCs) are responsible for tumor initiation, drug resistance, metastasis, and recurrence. In this study, we explored the role of CD146 in the regulation of liver CSCs. Here, we demonstrated that CD146 was highly expressed in liver CSCs. CD146 overexpression promoted the self-renewal ability and chemoresistance of Hepatocellular Carcinoma (HCC) cells in vitro and tumorigenicity in vivo. Inversely, knockdown of CD146 restrained these abilities. Mechanistically, CD146 activated the NF-κB signaling to up-regulate JAG2 expression and activated the Notch signaling, which resulted in increased stemness of HCC. Furthermore, JAG2 overexpression restored the Notch signaling activity, the stemness, and chemotherapeutic resistance caused by CD146 knockdown. These results demonstrated that CD146 positively regulates HCC stemness by activating the JAG2-NOTCH signaling. Combined targeting of CD146 and JAG2 may represent a novel therapeutic strategy for HCC treatment.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/drug therapy
- Humans
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/drug therapy
- Jagged-2 Protein/metabolism
- Jagged-2 Protein/genetics
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/drug effects
- Signal Transduction
- Receptors, Notch/metabolism
- Drug Resistance, Neoplasm/genetics
- Animals
- CD146 Antigen/metabolism
- CD146 Antigen/genetics
- Mice
- Cell Line, Tumor
- Mice, Nude
- Gene Expression Regulation, Neoplastic
- Mice, Inbred BALB C
- Male
Collapse
Affiliation(s)
- Bing Yan
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
- Department of General Surgery, Pingxiang People's Hospital, Pingxiang, 337000, China
| | - QiuYu Lu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - TianMing Gao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - KunQing Xiao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - QianNi Zong
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - HongWei Lv
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - GuiShuai Lv
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Liang Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - ChunYing Liu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China.
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China.
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology, Shanghai, 200438, China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - Wen Yang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China.
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China.
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology, Shanghai, 200438, China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - GuoQing Jiang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China.
| |
Collapse
|
3
|
Gurriaran-Rodriguez U, Datzkiw D, Radusky LG, Esper M, Javandoost E, Xiao F, Ming H, Fisher S, Marina A, De Repentigny Y, Kothary R, Azkargorta M, Elortza F, Rojas AL, Serrano L, Hierro A, Rudnicki MA. Identification of the Wnt signal peptide that directs secretion on extracellular vesicles. SCIENCE ADVANCES 2024; 10:eado5914. [PMID: 39661666 PMCID: PMC11633749 DOI: 10.1126/sciadv.ado5914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024]
Abstract
Wnt proteins are hydrophobic glycoproteins that are nevertheless capable of long-range signaling. We found that Wnt7a is secreted long distance on the surface of extracellular vesicles (EVs) following muscle injury. We defined a signal peptide region in Wnts required for secretion on EVs, termed exosome-binding peptide (EBP). Addition of EBP to an unrelated protein directed secretion on EVs. Palmitoylation and the signal peptide were not required for Wnt7a-EV secretion. Coatomer was identified as the EV-binding protein for the EBP. Analysis of cocrystal structures, binding thermodynamics, and mutagenesis found that a dilysine motif mediates EBP binding to coatomer with a conserved function across the Wnt family. We showed that EBP is required for Wnt7a bioactivity when expressed in vivo during regeneration. Overall, our study has elucidated the structural basis and singularity of Wnt secretion on EVs, alternatively to canonical secretion, opening avenues for innovative therapeutic targeting strategies and systemic protein delivery.
Collapse
Affiliation(s)
- Uxia Gurriaran-Rodriguez
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David Datzkiw
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Leandro G. Radusky
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Marie Esper
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ehsan Javandoost
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Fan Xiao
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
| | - Hong Ming
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
| | - Solomon Fisher
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alberto Marina
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Yves De Repentigny
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
| | - Rashmi Kothary
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Mikel Azkargorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Felix Elortza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Adriana L. Rojas
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Luis Serrano
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Aitor Hierro
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Michael A. Rudnicki
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Wu Z, Zang Y, Li C, He Z, Liu J, Du Z, Ma X, Jing L, Duan H, Feng J, Yan X. CD146, a therapeutic target involved in cell plasticity. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1563-1578. [PMID: 38613742 DOI: 10.1007/s11427-023-2521-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/28/2023] [Indexed: 04/15/2024]
Abstract
Since its identification as a marker for advanced melanoma in the 1980s, CD146 has been found to have multiple functions in both physiological and pathological processes, including embryonic development, tissue repair and regeneration, tumor progression, fibrosis disease, and inflammations. Subsequent research has revealed that CD146 is involved in various signaling pathways as a receptor or co-receptor in these processes. This correlation between CD146 and multiple diseases has sparked interest in its potential applications in diagnosis, prognosis, and targeted therapy. To better comprehend the versatile roles of CD146, we have summarized its research history and synthesized findings from numerous reports, proposing that cell plasticity serves as the underlying mechanism through which CD146 contributes to development, regeneration, and various diseases. Targeting CD146 would consequently halt cell state shifting during the onset and progression of these related diseases. Therefore, the development of therapy targeting CD146 holds significant practical value.
Collapse
Affiliation(s)
- Zhenzhen Wu
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuzhe Zang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chuyi Li
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhiheng He
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jingyu Liu
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhaoqi Du
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinran Ma
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lin Jing
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hongxia Duan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China.
| | - Jing Feng
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China.
- Joint Laboratory of Nanozymes in Zhengzhou University, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
5
|
Peng L, Zhang D, Tu H, Wu D, Xiang S, Yang W, Zhao Y, Yang J. The role of Map1b in regulating osteoblast polarity, proliferation, differentiation and migration. Bone 2024; 181:117038. [PMID: 38316337 DOI: 10.1016/j.bone.2024.117038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/07/2024]
Abstract
Osteoblast polarity, proliferation, differentiation, and migration are essential for maintaining normal bone structure and function. While the microtubule-associated protein Map1b has been extensively studied in nerve cells, its role in bone cells is less known. We investigated the functional significance of Map1b in mouse bone marrow stromal cells (ST2) and elucidated its relationship and influence on cytoskeletal polarity and Golgi organization. Our results suggest that Map1b, as a microtubule regulatory protein, can also regulate the expression of cyclin PCNA, p-H3(S10) and migration-related protein integrin β1, thereby affecting the proliferation and migration of osteoblasts. The downstream target gene Rgc32 was screened by RNA sequencing. Furthermore, Map1b, as a downstream mediator, regulates the Wnt5a signaling pathway. This study expands our understanding of the involvement of Map1b in bone biology and highlights its crucial role in governing osteoblast polarity, proliferation, and migration, thereby providing a basis for developing novel therapeutic strategies targeting Map1b in orthopedic medicine and promoting precision treatment modalities. Further investigations on the precise mechanisms underlying Map1b's influence on bone cell function and disease progression are needed.
Collapse
Affiliation(s)
- Li Peng
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Heng Tu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Dan Wu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shuaixi Xiang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenbin Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yun Zhao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
6
|
Mouawad N, Ruggeri E, Capasso G, Martinello L, Visentin A, Frezzato F, Trentin L. How receptor tyrosine kinase-like orphan receptor 1 meets its partners in chronic lymphocytic leukemia. Hematol Oncol 2024; 42:e3250. [PMID: 38949887 DOI: 10.1002/hon.3250] [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: 10/12/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 07/03/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is the most common leukemia in western societies, recognized by clinical and molecular heterogeneity. Despite the success of targeted therapies, acquired resistance remains a challenge for relapsed and refractory CLL, as a consequence of mutations in the target or the upregulation of other survival pathways leading to the progression of the disease. Research on proteins that can trigger such pathways may define novel therapies for a successful outcome in CLL such as the receptor tyrosine kinase-like orphan receptor 1 (ROR1). ROR1 is a signaling receptor for Wnt5a, with an important role during embryogenesis. The aberrant expression on CLL cells and several types of tumors, is involved in cell proliferation, survival, migration as well as drug resistance. Antibody-based immunotherapies and small-molecule compounds emerged to target ROR1 in preclinical and clinical studies. Efforts have been made to identify new prognostic markers having predictive value to refine and increase the detection and management of CLL. ROR1 can be considered as an attractive target for CLL diagnosis, prognosis, and treatment. It can be clinically effective alone and/or in combination with current approved agents. In this review, we summarize the scientific achievements in targeting ROR1 for CLL diagnosis, prognosis, and treatment.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Humans
- Receptor Tyrosine Kinase-like Orphan Receptors/metabolism
- Prognosis
- Molecular Targeted Therapy
- Animals
- Biomarkers, Tumor/metabolism
Collapse
Affiliation(s)
- Nayla Mouawad
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Edoardo Ruggeri
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Guido Capasso
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Leonardo Martinello
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Andrea Visentin
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Federica Frezzato
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Livio Trentin
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| |
Collapse
|
7
|
Miller SG, Hoh M, Ebmeier CC, Tay JW, Ahn NG. Cooperative polarization of MCAM/CD146 and ERM family proteins in melanoma. Mol Biol Cell 2024; 35:ar31. [PMID: 38117590 PMCID: PMC10916866 DOI: 10.1091/mbc.e23-06-0255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/22/2023] [Accepted: 12/15/2023] [Indexed: 12/22/2023] Open
Abstract
The WRAMP structure is a protein network associated with tail-end actomyosin contractility, membrane retraction, and directional persistence during cell migration. A marker of WRAMP structures is melanoma cell adhesion molecule (MCAM) which dynamically polarizes to the cell rear. However, factors that mediate MCAM polarization are still unknown. In this study, BioID using MCAM as bait identifies the ERM family proteins, moesin, ezrin, and radixin, as WRAMP structure components. We also present a novel image analysis pipeline, Protein Polarity by Percentile ("3P"), which classifies protein polarization using machine learning and facilitates quantitative analysis. Using 3P, we find that depletion of moesin, and to a lesser extent ezrin, decreases the proportion of cells with polarized MCAM. Furthermore, although copolarized MCAM and ERM proteins show high spatial overlap, 3P identifies subpopulations with ERM proteins closer to the cell periphery. Live-cell imaging confirms that MCAM and ERM protein polarization is tightly coordinated, but ERM proteins enrich at the cell edge first. Finally, deletion of a juxtamembrane segment in MCAM previously shown to promote ERM protein interactions impedes MCAM polarization. Our findings highlight the requirement for ERM proteins in recruitment of MCAM to WRAMP structures and an advanced computational tool to characterize protein polarization.
Collapse
Affiliation(s)
- Suzannah G. Miller
- Department of Biochemistry, University of Colorado Boulder, Boulder CO 80303
| | - Maria Hoh
- Department of Biochemistry, University of Colorado Boulder, Boulder CO 80303
| | | | - Jian Wei Tay
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO 80303
| | - Natalie G. Ahn
- Department of Biochemistry, University of Colorado Boulder, Boulder CO 80303
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO 80303
| |
Collapse
|
8
|
Ma YQ, Zhang XY, Zhao SW, Li D, Cai MQ, Yang H, Wang XM, Xue H. Retinoic acid delays murine palatal shelf elevation by inhibiting Wnt5a-mediated noncanonical Wnt signaling and downstream Cdc-42/F-actin remodeling in mesenchymal cells. Birth Defects Res 2023; 115:1658-1673. [PMID: 37675882 DOI: 10.1002/bdr2.2244] [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: 03/03/2023] [Revised: 08/12/2023] [Accepted: 08/21/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Mammalian palatal shelves erupted from maxillary prominences undergo vertical extention, transient elevation, and horizontal growth to fuse. Previous studies in mice reported that the retinoic acid (RA) contributed to cleft palate in high incidence by delaying the elevating procedure, but little was known about the underlying biological mechanisms. METHODS In this study, hematoxylin-eosin and immunofluorescence staining were employed to evaluate the phenotypes and the expression of related markers in the RA-treated mice model. In situ hybridization and RT-qPCR were used to detect the expression of genes involved in Wnt signaling pathway. The palatal mesenchymal cells were cultured in vitro, and stimulated with RA or CASIN, and co-treated with Foxy5. Wnt5a and Ccd42 expression were evaluated by immunofluorescence staining. Phalloidin was used to label the microfilament cytoskeleton (F-actin) in cultured cells. RESULTS We revealed that RA resulted in 100% incidence of cleft palate in mouse embryos, and the expression of genes responsible for Wnt5a-mediated noncanonical Wnt signal transduction were specifically downregulated in mesenchymal palatal shelves. The in vitro study of palatal mesenchymal cells indicated that RA treatment disrupted the organized remodeling of cytoskeleton, an indicative structure of cell migration regulated by the small Rho GTPase Cdc42. Moreover, we showed that the suppression of cytoskeleton and cell migration induced by RA was partially restored using the small molecule Foxy-5-mediated activation of Wnt5A, and this restoration was attenuated by CASIN (a selective GTPase Cdc42 inhibitor) again. CONCLUSIONS These data identified a crucial mechanism for Wnt5a-mediated noncanonical Wnt signaling in acting downstream of Rho GTPase Cdc42 to regulate cytoskeletal remodeling and cell migration during the process of palate elevation. Our study provided a new explanation for the cause of cleft palate induced by RA.
Collapse
Affiliation(s)
- Yan-Qing Ma
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing (No: 20JR10RA653 - ZDKF20210401), School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
- Department of Orthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Xin-Yu Zhang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing (No: 20JR10RA653 - ZDKF20210401), School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
- Department of Orthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Shi-Wei Zhao
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing (No: 20JR10RA653 - ZDKF20210401), School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
- Department of Orthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Dou Li
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing (No: 20JR10RA653 - ZDKF20210401), School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
- Department of Orthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Min-Qin Cai
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing (No: 20JR10RA653 - ZDKF20210401), School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
- Department of Orthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Hui Yang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing (No: 20JR10RA653 - ZDKF20210401), School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
- Department of Orthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Xiao-Ming Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing (No: 20JR10RA653 - ZDKF20210401), School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
- Department of Orthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Hui Xue
- Department of Stomatology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, People's Republic of China
| |
Collapse
|
9
|
Gurriaran-Rodriguez U, Datzkiw D, Radusky LG, Esper M, Xiao F, Ming H, Fisher S, Rojas MA, De Repentigny Y, Kothary R, Rojas AL, Serrano L, Hierro A, Rudnicki MA. Wnt binding to Coatomer proteins directs secretion on exosomes independently of palmitoylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542914. [PMID: 37398399 PMCID: PMC10312507 DOI: 10.1101/2023.05.30.542914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Wnt proteins are secreted hydrophobic glycoproteins that act over long distances through poorly understood mechanisms. We discovered that Wnt7a is secreted on extracellular vesicles (EVs) following muscle injury. Structural analysis identified the motif responsible for Wnt7a secretion on EVs that we term the Exosome Binding Peptide (EBP). Addition of the EBP to an unrelated protein directed secretion on EVs. Disruption of palmitoylation, knockdown of WLS, or deletion of the N-terminal signal peptide did not affect Wnt7a secretion on purified EVs. Bio-ID analysis identified Coatomer proteins as candidates responsible for loading Wnt7a onto EVs. The crystal structure of EBP bound to the COPB2 coatomer subunit, the binding thermodynamics, and mutagenesis experiments, together demonstrate that a dilysine motif in the EBP mediates binding to COPB2. Other Wnts contain functionally analogous structural motifs. Mutation of the EBP results in a significant impairment in the ability of Wnt7a to stimulate regeneration, indicating that secretion of Wnt7a on exosomes is critical for normal regeneration in vivo . Our studies have defined the structural mechanism that mediates binding of Wnt7a to exosomes and elucidated the singularity of long-range Wnt signalling.
Collapse
|
10
|
Abstract
In light of the demonstrated antagonism of Wnt5A signaling toward the growth of several bacterial pathogens, it was important to study the influence of Wnt5A on gut-resident bacteria and its outcome. Here, we demonstrate that in contrast to inhibiting the survival of the established gut pathogen Salmonella enterica, Wnt5A clearly promotes the survival of the common gut commensals Enterococcus faecalis and Lactobacillus rhamnosus within macrophages through a self-perpetuating Wnt5A-actin axis. A Wnt5A-actin axis furthermore regulates the subsistence of the natural bacterial population of the Peyer's patches, as is evident from the diminution in the countable bacterial CFU therein through the application of Wnt5A signaling and actin assembly inhibitors. Wnt5A dependency of the gut-resident bacterial population is also manifested in the notable difference between the bacterial diversities associated with the feces and Peyer's patches of Wnt5A heterozygous mice, which lack a functional copy of the Wnt5A gene, and their wild-type counterparts. Alterations in the gut commensal bacterial population resulting from either the lack of a copy of the Wnt5A gene or inhibitor-mediated attenuation of Wnt5A signaling are linked with significant differences in cell surface major histocompatibility complex (MHC) II levels and regulatory versus activated CD4 T cells associated with the Peyer's patches. Taken together, our findings reveal the significance of steady state Wnt5A signaling in shaping the gut commensal bacterial population and the T cell repertoire linked to it, thus unveiling a crucial control device for the maintenance of gut bacterial diversity and T cell homeostasis. IMPORTANCE Gut commensal bacterial diversity and T cell homeostasis are crucial entities of the host innate immune network, yet the molecular details of host-directed signaling pathways that sustain the steady state of gut bacterial colonization and T cell activation remain unclear. Here, we describe the protective role of a Wnt5A-actin axis in the survival of several gut bacterial commensals and its necessity in shaping gut bacterial colonization and the associated T cell repertoire. This study opens up new avenues of investigation into the role of the Wnt5A-actin axis in protection of the gut from dysbiosis-related inflammatory disorders.
Collapse
|
11
|
Zhang W, Yu J, Fu G, Li J, Huang H, Liu J, Yu D, Qiu M, Li F. ISL1/SHH/CXCL12 signaling regulates myogenic cell migration during mouse tongue development. Development 2022; 149:277065. [DOI: 10.1242/dev.200788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 09/15/2022] [Indexed: 11/06/2022]
Abstract
ABSTRACT
Migration of myoblasts derived from the occipital somites is essential for tongue morphogenesis. However, the molecular mechanisms of myoblast migration remain elusive. In this study, we report that deletion of Isl1 in the mouse mandibular epithelium leads to aglossia due to myoblast migration defects. Isl1 regulates the expression pattern of chemokine ligand 12 (Cxcl12) in the first branchial arch through the Shh/Wnt5a cascade. Cxcl12+ mesenchymal cells in Isl1ShhCre embryos were unable to migrate to the distal region, but instead clustered in a relatively small proximal domain of the mandible. CXCL12 serves as a bidirectional cue for myoblasts expressing its receptor CXCR4 in a concentration-dependent manner, attracting Cxcr4+ myoblast invasion at low concentrations but repelling at high concentrations. The accumulation of Cxcl12+ mesenchymal cells resulted in high local concentrations of CXCL12, which prevented Cxcr4+ myoblast invasion. Furthermore, transgenic activation of Ihh alleviated defects in tongue development and rescued myoblast migration, confirming the functional involvement of Hedgehog signaling in tongue development. In summary, this study provides the first line of genetic evidence that the ISL1/SHH/CXCL12 axis regulates myoblast migration during tongue development.
Collapse
Affiliation(s)
- Wei Zhang
- Zhejiang Key Laboratory 1 , Hangzhou 311121 , People's Republic of China
- of Organ Development and Regeneration, Department of Biological Sciences, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University 1 , Hangzhou 311121 , People's Republic of China
| | - Jiaojiao Yu
- Zhejiang Key Laboratory 1 , Hangzhou 311121 , People's Republic of China
- of Organ Development and Regeneration, Department of Biological Sciences, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University 1 , Hangzhou 311121 , People's Republic of China
| | - Guoquan Fu
- Zhejiang Key Laboratory 1 , Hangzhou 311121 , People's Republic of China
- of Organ Development and Regeneration, Department of Biological Sciences, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University 1 , Hangzhou 311121 , People's Republic of China
| | - Jianying Li
- Zhejiang Key Laboratory 1 , Hangzhou 311121 , People's Republic of China
- of Organ Development and Regeneration, Department of Biological Sciences, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University 1 , Hangzhou 311121 , People's Republic of China
| | - Huarong Huang
- Zhejiang Key Laboratory 1 , Hangzhou 311121 , People's Republic of China
- of Organ Development and Regeneration, Department of Biological Sciences, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University 1 , Hangzhou 311121 , People's Republic of China
| | - Jing Liu
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, Department of Environmental Sciences, College of Environmental and Resource Sciences, Zhejiang University 2 , Hangzhou 310058 , People's Republic of China
| | - Dongliang Yu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University 3 , Hangzhou 310018 , People's Republic of China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory 1 , Hangzhou 311121 , People's Republic of China
- of Organ Development and Regeneration, Department of Biological Sciences, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University 1 , Hangzhou 311121 , People's Republic of China
| | - Feixue Li
- Zhejiang Key Laboratory 1 , Hangzhou 311121 , People's Republic of China
- of Organ Development and Regeneration, Department of Biological Sciences, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University 1 , Hangzhou 311121 , People's Republic of China
| |
Collapse
|
12
|
Sarraf TR, Sen M. Wnt5A signaling supports antigen processing and CD8 T cell activation. Front Immunol 2022; 13:960060. [PMID: 36091060 PMCID: PMC9459031 DOI: 10.3389/fimmu.2022.960060] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Antigen processing and antigen-specific CD8 T cell activation form part and parcel of cell-mediated immunity to infections. Yet, several lacunae remain in our understanding of how antigen processing and CD8 T cell response are coordinated. In this study, using mouse bone marrow-derived dendritic cells (BMDC) as antigen-presenting cells and Ovalbumin (OVA)/DQ-Ovalbumin (DQ-OVA) as model antigen we demonstrated that Wnt5A signaling in BMDC supports antigen processing/presentation and concomitant CD8 T cell activation through regulation of actin and proteasome dynamics. Recombinant Wnt5A conditioning of BMDC and associated actin assembly facilitated DQ-OVA processing, which was inhibited by the proteasome inhibitor MG132. Moreover, Wnt5A depletion led to a significant reduction in OVA processing and presentation. Impaired DQ-OVA processing in Wnt5A depleted BMDC correlated with altered dynamics of both actin and the proteasome regulator PA28α-PA28β, and reduced association of DQ-OVA with actin and proteasome subunits. Inhibited OVA processing/presentation in the Wnt5A depleted BMDC also resulted in subdued activation of OVA-sensitized CD8 T cells in co-culture with the BMDC. In concurrence with these findings, we demonstrated reduced OVA processing and impaired CD8 T cell response to OVA immunization in Wnt5A heterozygous mice lacking a copy of the Wnt5A gene in comparison to the wild-type cohorts. Taken together, our results reveal a crucial requirement of Wnt5A signaling in antigen processing/presentation and CD8 T cell activation, thus unveiling a vital regulatory node of cell-mediated immunity, unidentified thus far.
Collapse
|
13
|
Hettinger ZR, Wen Y, Peck BD, Hamagata K, Confides AL, Van Pelt DW, Harrison DA, Miller BF, Butterfield TA, Dupont-Versteegden EE. Mechanotherapy Reprograms Aged Muscle Stromal Cells to Remodel the Extracellular Matrix during Recovery from Disuse. FUNCTION 2022; 3:zqac015. [PMID: 35434632 PMCID: PMC9009398 DOI: 10.1093/function/zqac015] [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: 01/22/2022] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 01/07/2023] Open
Abstract
Aging is accompanied by reduced remodeling of skeletal muscle extracellular matrix (ECM), which is exacerbated during recovery following periods of disuse atrophy. Mechanotherapy has been shown to promote ECM remodeling through immunomodulation in adult muscle recovery, but not during the aged recovery from disuse. In order to determine if mechanotherapy promotes ECM remodeling in aged muscle, we performed single cell RNA sequencing (scRNA-seq) of all mononucleated cells in adult and aged rat gastrocnemius muscle recovering from disuse, with (REM) and without mechanotherapy (RE). We show that fibroadipogenic progenitor cells (FAPs) in aged RE muscle are highly enriched in chemotaxis genes (Csf1), but absent in ECM remodeling genes compared to adult RE muscle (Col1a1). Receptor-ligand (RL) network analysis of all mononucleated cell populations in aged RE muscle identified chemotaxis-enriched gene expression in numerous stromal cell populations (FAPs, endothelial cells, pericytes), despite reduced enrichment of genes related to phagocytic activity in myeloid cell populations (macrophages, monocytes, antigen presenting cells). Following mechanotherapy, aged REM mononuclear cell gene expression resembled adult RE muscle as evidenced by RL network analyses and KEGG pathway activity scoring. To validate our transcriptional findings, ECM turnover was measured in an independent cohort of animals using in vivo isotope tracing of intramuscular collagen and histological scoring of the ECM, which confirmed mechanotherapy-mediated ECM remodeling in aged RE muscle. Our results highlight age-related cellular mechanisms underpinning the impairment to complete recovery from disuse, and also promote mechanotherapy as an intervention to enhance ECM turnover in aged muscle recovering from disuse.
Collapse
Affiliation(s)
- Zachary R Hettinger
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Yuan Wen
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Bailey D Peck
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Kyoko Hamagata
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Amy L Confides
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Douglas W Van Pelt
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Douglas A Harrison
- Department of Biology, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Timothy A Butterfield
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky; Lexington, KY 40536, USA
| | - Esther E Dupont-Versteegden
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
14
|
Afzal MW, Duan K, Zhang Y, Gao Y, Qin B, Wang G, Lei L, Tang H, Guo Y. A rhodol-based fluorescent probe with a pair of hydrophilic and rotatable wings for sensitively monitoring intracellular polarity. Chem Asian J 2022; 17:e202200063. [PMID: 35191194 DOI: 10.1002/asia.202200063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/21/2022] [Indexed: 11/06/2022]
Abstract
Cell polarity, as a vital intracellular microenvironment characteristic, has immense effects on numerous pathological and biological processes. Therefore, the tracking of polarity variations is highly essential to explore the role and mechanism of the polarity in pathophysiological processes. Herein, we designed and synthesized a novel rhodol-based fluorescent probe RDS sensitive to polarity by introducing a bis(2-hydroxyethylthio)methyl group, like a pair of hydrophilic and rotatable wings, into the rhodol skeleton. This unique design makes RDS adopt the colorless and non-fluorescent spirocyclic form in low polarity medium while the colored and fluorescent ring-open form in high polarity system, resulting in a positive-correlation response of fluorescence intension to polarity. Importantly, RDS was successfully applied to monitor the polarity changes in living cells including cancer cells, healthy cells and senescent healthy cells, visualizing that the polarity of cancer cells is lower than that of healthy cells in which the more senescent ones have higher polarity.
Collapse
Affiliation(s)
| | - Kaixuan Duan
- Northwest University, College of Chemistry and Materials Science, CHINA
| | - Yanhui Zhang
- Northwest University, College of Chemistry and Materials Science, CHINA
| | - Ying Gao
- Northwest University, College of Chemistry and Materials Science, CHINA
| | - Bo Qin
- Xi'an University of Posts and Telecommunications, School of Automation, CHINA
| | - Guangwei Wang
- Northwest University, College of Chemistry and Materials Science, CHINA
| | - Lin Lei
- Northwest University, College of Chemistry and Materials Science, CHINA
| | - Haoyang Tang
- Xi'an University of Posts and Telecommunications, School of Automation, CHINA
| | - Yuan Guo
- Northwest University, College of Chemistry and Materials Science, 1 Xuefu Road, Chang'an district, 710127, Xi'an, CHINA
| |
Collapse
|
15
|
Čada Š, Bryja V. Local Wnt signalling in the asymmetric migrating vertebrate cells. Semin Cell Dev Biol 2021; 125:26-36. [PMID: 34896020 DOI: 10.1016/j.semcdb.2021.11.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/27/2022]
Abstract
Wnt signalling is known to generate cellular asymmetry via Wnt/planar cell polarity pathway (Wnt/PCP). Wnt/PCP acts locally (i) to orient membrane polarity and asymmetric establishment of intercellular junctions via conserved set of PCP proteins most specifically represented by Vangl and Prickle, and (ii) to asymmetrically rearrange cytoskeletal structures via downstream effectors of Dishevelled (Dvl). This process is best described on stable phenotypes of epithelial cells. Here, however, we review the activity of Wnt signalling in migratory cells which experience the extensive rearrangements of cytoskeleton and consequently dynamic asymmetry, making the localised effects of Wnt signalling easier to distinguish. Firstly, we focused on migration of neuronal axons, which allows to study how the pre-existent cellular asymmetry can influence Wnt signalling outcome. Then, we reviewed the role of Wnt signalling in models of mesenchymal migration including neural crest, melanoma, and breast cancer cells. Last, we collected evidence for local Wnt signalling in amoeboid cells, especially lymphocytes. As the outcome of this review, we identify blank spots in our current understanding of this topic, propose models that synthesise the current observations and allow formulation of testable hypotheses for the future research.
Collapse
Affiliation(s)
- Štěpán Čada
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; Department of Cytokinetics, Institute of Biophysics CAS, Královopolská 135, 61265 Brno, Czech Republic.
| |
Collapse
|
16
|
Astudillo P. An emergent Wnt5a/YAP/TAZ regulatory circuit and its possible role in cancer. Semin Cell Dev Biol 2021; 125:45-54. [PMID: 34764023 DOI: 10.1016/j.semcdb.2021.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 12/29/2022]
Abstract
Wnt5a is a ligand that plays several roles in development, homeostasis, and disease. A growing body of evidence indicates that Wnt5a is involved in cancer progression. Despite extensive research in this field, our knowledge about how Wnt5a is precisely involved in cancer is still incomplete. It is usually thought that certain combinations of Frizzled receptors and co-receptors might explain the observed effects of Wnt5a either as a tumor suppressor or by promoting migration and invasion. While accepting this 'receptor context' model, this review proposes that Wnt5a is integrated within a larger regulatory circuit involving β-catenin, YAP/TAZ, and LATS1/2. Remarkably, WNT5A and YAP1 are transcriptionally regulated by the Hippo and Wnt pathways, respectively, and might form a regulatory circuit acting through LATS kinases and secreted Wnt/β-catenin inhibitors, including Wnt5a itself. Therefore, understanding the precise role of Wnt5a and YAP in cancer requires a systems biology perspective.
Collapse
Affiliation(s)
- Pablo Astudillo
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
17
|
Aghaizu ND, Warre-Cornish KM, Robinson MR, Waldron PV, Maswood RN, Smith AJ, Ali RR, Pearson RA. Repeated nuclear translocations underlie photoreceptor positioning and lamination of the outer nuclear layer in the mammalian retina. Cell Rep 2021; 36:109461. [PMID: 34348137 PMCID: PMC8356022 DOI: 10.1016/j.celrep.2021.109461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 11/19/2019] [Accepted: 07/09/2021] [Indexed: 12/28/2022] Open
Abstract
In development, almost all stratified neurons must migrate from their birthplace to the appropriate neural layer. Photoreceptors reside in the most apical layer of the retina, near their place of birth. Whether photoreceptors require migratory events for fine-positioning and/or retention within this layer is not well understood. Here, we show that photoreceptor nuclei of the developing mouse retina cyclically exhibit rapid, dynein-1-dependent translocation toward the apical surface, before moving more slowly in the basal direction, likely due to passive displacement by neighboring retinal nuclei. Attenuating dynein 1 function in rod photoreceptors results in their ectopic basal displacement into the outer plexiform layer and inner nuclear layer. Synapse formation is also compromised in these displaced cells. We propose that repeated, apically directed nuclear translocation events are necessary to ensure retention of post-mitotic photoreceptors within the emerging outer nuclear layer during retinogenesis, which is critical for correct neuronal lamination.
Collapse
Affiliation(s)
- Nozie D Aghaizu
- University College London Institute of Ophthalmology, London EC1V 9EL, UK.
| | | | - Martha R Robinson
- University College London Institute of Ophthalmology, London EC1V 9EL, UK
| | - Paul V Waldron
- University College London Institute of Ophthalmology, London EC1V 9EL, UK
| | - Ryea N Maswood
- University College London Institute of Ophthalmology, London EC1V 9EL, UK
| | - Alexander J Smith
- University College London Institute of Ophthalmology, London EC1V 9EL, UK; Centre for Cell and Gene Therapy, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Robin R Ali
- University College London Institute of Ophthalmology, London EC1V 9EL, UK; Centre for Cell and Gene Therapy, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Rachael A Pearson
- University College London Institute of Ophthalmology, London EC1V 9EL, UK; Centre for Cell and Gene Therapy, King's College London, Guy's Hospital, London SE1 9RT, UK.
| |
Collapse
|
18
|
Chavkin NW, Sano S, Wang Y, Oshima K, Ogawa H, Horitani K, Sano M, MacLauchlan S, Nelson A, Setia K, Vippa T, Watanabe Y, Saucerman JJ, Hirschi KK, Gokce N, Walsh K. The Cell Surface Receptors Ror1/2 Control Cardiac Myofibroblast Differentiation. J Am Heart Assoc 2021; 10:e019904. [PMID: 34155901 PMCID: PMC8403294 DOI: 10.1161/jaha.120.019904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/22/2021] [Indexed: 12/25/2022]
Abstract
Background A hallmark of heart failure is cardiac fibrosis, which results from the injury-induced differentiation response of resident fibroblasts to myofibroblasts that deposit extracellular matrix. During myofibroblast differentiation, fibroblasts progress through polarization stages of early proinflammation, intermediate proliferation, and late maturation, but the regulators of this progression are poorly understood. Planar cell polarity receptors, receptor tyrosine kinase-like orphan receptor 1 and 2 (Ror1/2), can function to promote cell differentiation and transformation. In this study, we investigated the role of the Ror1/2 in a model of heart failure with emphasis on myofibroblast differentiation. Methods and Results The role of Ror1/2 during cardiac myofibroblast differentiation was studied in cell culture models of primary murine cardiac fibroblast activation and in knockout mouse models that underwent transverse aortic constriction surgery to induce cardiac injury by pressure overload. Expression of Ror1 and Ror2 were robustly and exclusively induced in fibroblasts in hearts after transverse aortic constriction surgery, and both were rapidly upregulated after early activation of primary murine cardiac fibroblasts in culture. Cultured fibroblasts isolated from Ror1/2 knockout mice displayed a proinflammatory phenotype indicative of impaired myofibroblast differentiation. Although the combined ablation of Ror1/2 in mice did not result in a detectable baseline phenotype, transverse aortic constriction surgery led to the death of all mice by day 6 that was associated with myocardial hyperinflammation and vascular leakage. Conclusions Together, these results show that Ror1/2 are essential for the progression of myofibroblast differentiation and for the adaptive remodeling of the heart in response to pressure overload.
Collapse
Affiliation(s)
- Nicholas W. Chavkin
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Cell BiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Soichi Sano
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
- Department of CardiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Ying Wang
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Kosei Oshima
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Hayato Ogawa
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Keita Horitani
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Miho Sano
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Susan MacLauchlan
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Anders Nelson
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of PharmacologyUniversity of VirginiaCharlottesvilleVA
| | - Karishma Setia
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Tanvi Vippa
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Yosuke Watanabe
- Vascular Biology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Jeffrey J. Saucerman
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVA
| | - Karen K. Hirschi
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Cell BiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Cardiovascular Research CenterSchool of MedicineYale UniversityNew HavenCT
| | - Noyan Gokce
- Boston University School of MedicineBostonMA
| | - Kenneth Walsh
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| |
Collapse
|
19
|
Proteomic analysis identifies the E3 ubiquitin ligase Pdzrn3 as a regulatory target of Wnt5a-Ror signaling. Proc Natl Acad Sci U S A 2021; 118:2104944118. [PMID: 34135125 DOI: 10.1073/pnas.2104944118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Wnt5a-Ror signaling is a conserved pathway that regulates morphogenetic processes during vertebrate development [R. T. Moon et al, Development 119, 97-111 (1993); I. Oishi et al, Genes Cells 8, 645-654 (2003)], but its downstream signaling events remain poorly understood. Through a large-scale proteomic screen in mouse embryonic fibroblasts, we identified the E3 ubiquitin ligase Pdzrn3 as a regulatory target of the Wnt5a-Ror pathway. Upon pathway activation, Pdzrn3 is degraded in a β-catenin-independent, ubiquitin-proteasome system-dependent manner. We developed a flow cytometry-based reporter to monitor Pdzrn3 abundance and delineated a signaling cascade involving Frizzled, Dishevelled, Casein kinase 1, and Glycogen synthase kinase 3 that regulates Pdzrn3 stability. Epistatically, Pdzrn3 is regulated independently of Kif26b, another Wnt5a-Ror effector. Wnt5a-dependent degradation of Pdzrn3 requires phosphorylation of three conserved amino acids within its C-terminal LNX3H domain [M. Flynn, O. Saha, P. Young, BMC Evol. Biol. 11, 235 (2011)], which acts as a bona fide Wnt5a-responsive element. Importantly, this phospho-dependent degradation is essential for Wnt5a-Ror modulation of cell migration. Collectively, this work establishes a Wnt5a-Ror cell morphogenetic cascade involving Pdzrn3 phosphorylation and degradation.
Collapse
|
20
|
Gai Z, Wang Y, Tian L, Gong G, Zhao J. Whole Genome Level Analysis of the Wnt and DIX Gene Families in Mice and Their Coordination Relationship in Regulating Cardiac Hypertrophy. Front Genet 2021; 12:608936. [PMID: 34168671 PMCID: PMC8217762 DOI: 10.3389/fgene.2021.608936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 05/17/2021] [Indexed: 12/27/2022] Open
Abstract
The Wnt signaling pathway is an evolutionarily conserved signaling pathway that plays essential roles in embryonic development, organogenesis, and many other biological activities. Both Wnt proteins and DIX proteins are important components of Wnt signaling. Systematic studies of Wnt and DIX families at the genome-wide level may provide a comprehensive landscape to elucidate their functions and demonstrate their relationships, but they are currently lacking. In this report, we describe the correlations between mouse Wnt and DIX genes in family expansion, molecular evolution, and expression levels in cardiac hypertrophy at the genome-wide scale. We observed that both the Wnt and DIX families underwent more expansion than the overall average in the evolutionarily early stage. In addition, mirrortree analyses suggested that Wnt and DIX were co-evolved protein families. Collectively, these results would help to elucidate the evolutionary characters of Wnt and DIX families and demonstrate their correlations in mediating cardiac hypertrophy.
Collapse
Affiliation(s)
- Zhongchao Gai
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Yujiao Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Lu Tian
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Guoli Gong
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Jieqiong Zhao
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| |
Collapse
|
21
|
Jobe NP, Åsberg L, Andersson T. Reduced WNT5A signaling in melanoma cells favors an amoeboid mode of invasion. Mol Oncol 2021; 15:1835-1848. [PMID: 33969605 PMCID: PMC8253101 DOI: 10.1002/1878-0261.12974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 02/25/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor cells invade and spread via either a mesenchymal or an amoeboid mode of migration. Amoeboid tumor cells have a rounded morphology and pronounced RhoA activity. Here, we investigate how WNT5A signaling, a tumor promotor in melanoma, relates to Rho GTPase activity and amoeboid migration. We compared melanoma cells with low (HTB63 cells) and high (WM852 cells) WNT5A expression. HTB63 cells exhibited an amoeboid morphology and had higher RhoA activity but lower invasiveness than WM852 cells in a three‐dimensional (3D) collagen matrix. We next explored the relationships between WNT5A, morphology, and invasive behavior. WNT5A knockdown impaired Rho GTPase Cdc42 activity, resulting in reduced invasion of amoeboid and mesenchymal melanoma cells. Interestingly, knockdown of WNT5A or inhibition of its secretion in WM852 cells expressing wild‐type BRAF also led to increased RhoA activity via decreased RND3 expression, resulting in predominantly amoeboid morphology. In contrast, such treatments had the opposite effects on RND3 expression and RhoA activity in HTB63 cells expressing the active BRAFV600 mutation. However, treatment of HTB63 cells with a BRAF inhibitor made them respond to WNT5A knockdown in a similar manner as WM852 cells expressing wild‐type BRAF. We next found that dual targeting of WNT5A and RhoA more effectively reduced melanoma cell invasion than targeting either protein individually. Taken together, our results suggest that low WNT5A signaling in melanoma cells promotes a rounded amoeboid type of invasion, which quite likely serves as a compensatory response to decreased WNT5A/Cdc42‐driven invasion. This phenomenon partially explains the enduring melanoma cell invasion observed after impaired WNT5A signaling and has therapeutic implications. Our results suggest that dual targeting of WNT5A and RhoA signaling is a more effective strategy for controlling the invasion of BRAF wild‐type and BRAFV600 mutated melanomas treated with a BRAF inhibitor than targeting either of the proteins individually.
Collapse
Affiliation(s)
- Njainday Pulo Jobe
- Experimental Pathology, Department of Translational Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Lisa Åsberg
- Experimental Pathology, Department of Translational Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Tommy Andersson
- Experimental Pathology, Department of Translational Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
22
|
Cheung SY, Sayeed M, Nakuluri K, Li L, Feldman BJ. MYH9 facilitates autoregulation of adipose tissue depot development. JCI Insight 2021; 6:136233. [PMID: 33986190 PMCID: PMC8262332 DOI: 10.1172/jci.insight.136233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/31/2021] [Indexed: 01/12/2023] Open
Abstract
White adipose tissue not only serves as a reservoir for energy storage but also secretes a variety of hormonal signals and modulates systemic metabolism. A substantial amount of adipose tissue develops in early postnatal life, providing exceptional access to the formation of this important tissue. Although a number of factors have been identified that can modulate the differentiation of progenitor cells into mature adipocytes in cell-autonomous assays, it remains unclear which are connected to physiological extracellular inputs and are most relevant to tissue formation in vivo. Here, we elucidate that mature adipocytes themselves signal to adipose depot–resident progenitor cells to direct depot formation in early postnatal life and gate adipogenesis when the tissue matures. Our studies revealed that as the adipose depot matures, a signal generated in mature adipocytes is produced, converges on progenitor cells to regulate the cytoskeletal protein MYH9, and attenuates the rate of adipogenesis in vivo.
Collapse
Affiliation(s)
| | | | | | | | - Brian J Feldman
- Department of Pediatrics.,Nutrition Obesity Research Center.,Diabetes Research Center, and.,Developmental and Stem Cell Biology Program, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
23
|
Rogan MR, Patterson LL, Byerly CD, Luo T, Paessler S, Veljkovic V, Quade B, McBride JW. Ehrlichia chaffeensis TRP120 Is a Wnt Ligand Mimetic That Interacts with Wnt Receptors and Contains a Novel Repetitive Short Linear Motif That Activates Wnt Signaling. mSphere 2021; 6:6/2/e00216-21. [PMID: 33883266 PMCID: PMC8546699 DOI: 10.1128/msphere.00216-21] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ehrlichia chaffeensis expresses the TRP120 multifunctional effector, which is known to play a role in phagocytic entry, on the surface of infectious dense-cored ehrlichiae, but a cognate host receptor has not been identified. We recently reported that E. chaffeensis activates canonical Wnt signaling in monocytes to promote bacterial uptake and intracellular survival and that TRP120 was involved in this activation event. To identify the specific mechanism of pathway activation, we hypothesized that TRP120 is a Wnt signaling ligand mimetic that initiates Wnt pathway activity through direct interaction with the Wnt pathway Frizzled family of receptors. In this study, we used confocal immunofluorescence microscopy to demonstrate very strong colocalization between E. chaffeensis and Fzd2, 4, 5, 7, and 9 as well as coreceptor LRP5 at 1 to 3 h postinfection. Direct binding between TRP120 and multiple Fzd receptors was further confirmed by enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR). Interfering RNA knockdown of Wnt receptors, coreceptors, and signaling pathway components significantly reduced E. chaffeensis infection, demonstrating that complex and redundant interactions are involved in Wnt pathway exploitation. We utilized in silico approaches to identify a repetitive short linear motif (SLiM) in TRP120 that is homologous to Wnt ligands and used mutant SLiM peptides and an α-TRP120-Wnt-SLiM antibody to demonstrate that the TRP120 Wnt SLiM activates the canonical Wnt pathway and promotes E. chaffeensis infection. This study reports the first example of bacterial mimicry of Wnt pathway ligands and highlights a pathogenic mechanism with potential for targeting by antimicrobial therapeutics.IMPORTANCE Upon infecting mammalian hosts, Ehrlichia chaffeensis establishes a replicative niche in microbe-eating immune system cells where it expertly orchestrates infection and spread. One of the ways Ehrlichia survives within these phagocytes is by activating evolutionarily conserved signaling pathways including the Wnt pathway; however, the molecular details of pathway hijacking have not been defined. This study is significant because it identifies an ehrlichial protein that directly interacts with components of the Wnt receptor complex, influencing pathway activity and promoting infection. Consequentially, Ehrlichia serves as a unique tool to investigate the intricacies of how pathogens repurpose human immune cell signaling and provides an opportunity to better understand many cellular processes in health and disease. Furthermore, understanding how this bacterium utilizes its small genome to survive within cells that evolved to destroy pathogens will facilitate the development of antibacterial therapeutics that could target Ehrlichia as well as other intracellular agents of human disease.
Collapse
Affiliation(s)
- Madison R Rogan
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - LaNisha L Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Caitlan D Byerly
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Tian Luo
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- BiomedProtection, LLC, Galveston, Texas, USA
| | | | - Bethany Quade
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jere W McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
24
|
Jati S, Sengupta S, Sen M. Wnt5A-Mediated Actin Organization Regulates Host Response to Bacterial Pathogens and Non-Pathogens. Front Immunol 2021; 11:628191. [PMID: 33664738 PMCID: PMC7921742 DOI: 10.3389/fimmu.2020.628191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/21/2020] [Indexed: 11/14/2022] Open
Abstract
Wnt5A signaling facilitates the killing of several bacterial pathogens, but not the non-pathogen E. coli DH5α. The basis of such pathogen vs. non-pathogen distinction is unclear. Accordingly, we analyzed the influence of Wnt5A signaling on pathogenic E. coli K1 in relation to non-pathogenic E. coli K12-MG1655 and E. coli DH5α eliminating interspecies variability from our study. Whereas cell internalized E. coli K1 disrupted cytoskeletal actin organization and multiplied during Wnt5A depletion, rWnt5A mediated activation revived cytoskeletal actin assembly facilitating K1 eradication. Cell internalized E. coli K12-MG1655 and E. coli DH5α, which did not perturb actin assembly appreciably, remained unaffected by rWnt5A treatment. Phagosomes prepared separately from Wnt5A conditioned medium treated K1 and K12-MG1655 infected macrophages revealed differences in the relative levels of actin and actin network promoting proteins, upholding that the Wnt5A-Actin axis operates differently for internalized pathogen and non-pathogen. Interestingly, exposure of rWnt5A treated K1 and K12-MG1655/DH5α infected macrophages to actin assembly inhibitors reversed the scenario, blocking killing of K1, yet promoting killing of both K12-MG1655 and DH5α. Taken together, our study illustrates that the state of activation of the Wnt5A/Actin axis in the context of the incumbent bacteria is crucial for directing host response to infection.
Collapse
Affiliation(s)
- Suborno Jati
- Division of Cancer Biology and Inflammatory Disorder, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Soham Sengupta
- Division of Cancer Biology and Inflammatory Disorder, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Malini Sen
- Division of Cancer Biology and Inflammatory Disorder, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
25
|
CD146/sCD146 in the Pathogenesis and Monitoring of Angiogenic and Inflammatory Diseases. Biomedicines 2020; 8:biomedicines8120592. [PMID: 33321883 PMCID: PMC7764286 DOI: 10.3390/biomedicines8120592] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/04/2020] [Indexed: 12/19/2022] Open
Abstract
CD146 is a cell adhesion molecule expressed on endothelial cells, as well as on other cells such as mesenchymal stem cells and Th17 lymphocytes. This protein also exists in a soluble form, whereby it can be detected in biological fluids, including the serum or the cerebrospinal fluid (CSF). Some studies have highlighted the significance of CD146 and its soluble form in angiogenesis and inflammation, having been shown to contribute to the pathogenesis of many inflammatory autoimmune diseases, such as systemic sclerosis, mellitus diabetes, rheumatoid arthritis, inflammatory bowel diseases, and multiple sclerosis. In this review, we will focus on how CD146 and sCD146 contribute to the pathogenesis of the aforementioned autoimmune diseases and discuss the relevance of considering it as a biomarker in these pathologies.
Collapse
|
26
|
Cheong SS, Akram KM, Matellan C, Kim SY, Gaboriau DCA, Hind M, del Río Hernández AE, Griffiths M, Dean CH. The Planar Polarity Component VANGL2 Is a Key Regulator of Mechanosignaling. Front Cell Dev Biol 2020; 8:577201. [PMID: 33195213 PMCID: PMC7658195 DOI: 10.3389/fcell.2020.577201] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 10/08/2020] [Indexed: 12/02/2022] Open
Abstract
VANGL2 is a component of the planar cell polarity (PCP) pathway, which regulates tissue polarity and patterning. The Vangl2 Lp mutation causes lung branching defects due to dysfunctional actomyosin-driven morphogenesis. Since the actomyosin network regulates cell mechanics, we speculated that mechanosignaling could be impaired when VANGL2 is disrupted. Here, we used live-imaging of precision-cut lung slices (PCLS) from Vangl2 Lp/+ mice to determine that alveologenesis is attenuated as a result of impaired epithelial cell migration. Vangl2 Lp/+ tracheal epithelial cells (TECs) and alveolar epithelial cells (AECs) exhibited highly disrupted actomyosin networks and focal adhesions (FAs). Functional assessment of cellular forces confirmed impaired traction force generation in Vangl2 Lp/+ TECs. YAP signaling in Vangl2 Lp airway epithelium was reduced, consistent with a role for VANGL2 in mechanotransduction. Furthermore, activation of RhoA signaling restored actomyosin organization in Vangl2 Lp/+ , confirming RhoA as an effector of VANGL2. This study identifies a pivotal role for VANGL2 in mechanosignaling, which underlies the key role of the PCP pathway in tissue morphogenesis.
Collapse
Affiliation(s)
- Sek-Shir Cheong
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Khondoker M. Akram
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Carlos Matellan
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Sally Yunsun Kim
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - David C. A. Gaboriau
- Facility for Imaging by Light Microscopy, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Matthew Hind
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- National Institute for Health Research, Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, London, United Kingdom
| | - Armando E. del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Mark Griffiths
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Peri-Operative Medicine Department, St Bartholomew’s Hospital, London, United Kingdom
| | - Charlotte H. Dean
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- MRC Harwell Institute, Harwell Campus, Oxfordshire, United Kingdom
| |
Collapse
|
27
|
Rao-Bhatia A, Zhu M, Yin WC, Coquenlorge S, Zhang X, Woo J, Sun Y, Dean CH, Liu A, Hui CC, Shivdasani RA, McNeill H, Hopyan S, Kim TH. Hedgehog-Activated Fat4 and PCP Pathways Mediate Mesenchymal Cell Clustering and Villus Formation in Gut Development. Dev Cell 2020; 52:647-658.e6. [PMID: 32155439 DOI: 10.1016/j.devcel.2020.02.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 11/06/2019] [Accepted: 02/03/2020] [Indexed: 12/16/2022]
Abstract
During development, intestinal epithelia undergo dramatic morphogenesis mediated by mesenchymal signaling to form villi, which are required for efficient nutrient absorption and host defense. Although both smooth-muscle-induced physical forces and mesenchymal cell clustering beneath emerging villi are implicated in epithelial folding, the underlying cellular mechanisms are unclear. Hedgehog (Hh) signaling can mediate both processes. We therefore analyzed its direct targetome and revealed GLI2 transcriptional activation of atypical cadherin and planar cell polarity (PCP) genes. By examining Fat4 and Dchs1 knockout mice, we demonstrate their critical roles in villus formation. Analyses of PCP-mutant mice and genetic interaction studies show that the Fat4-Dchs1 axis acts in parallel to the core-Vangl2 PCP axis to control mesenchymal cell clustering. Moreover, live light-sheet fluorescence microscopy and cultured PDGFRα+ cells reveal a requirement for PCP in their oriented cell migration guided by WNT5A. Therefore, mesenchymal PCP induced by Hh signaling drives cell clustering and subsequent epithelial remodeling.
Collapse
Affiliation(s)
- Abilasha Rao-Bhatia
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Min Zhu
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Wen-Chi Yin
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sabrina Coquenlorge
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xiaoyun Zhang
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Janghee Woo
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Charlotte H Dean
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Aimin Liu
- Department of Biology, Eberly College of Science, Centers for Cellular Dynamics and Molecular Investigation of Neurological Disorders, Huck Institute of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Chi-Chung Hui
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Helen McNeill
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sevan Hopyan
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tae-Hee Kim
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
28
|
Yu JJS, Maugarny-Calès A, Pelletier S, Alexandre C, Bellaiche Y, Vincent JP, McGough IJ. Frizzled-Dependent Planar Cell Polarity without Secreted Wnt Ligands. Dev Cell 2020; 54:583-592.e5. [PMID: 32888416 PMCID: PMC7497783 DOI: 10.1016/j.devcel.2020.08.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/07/2020] [Accepted: 08/10/2020] [Indexed: 12/26/2022]
Abstract
Planar cell polarity (PCP) organizes the orientation of cellular protrusions and migratory activity within the tissue plane. PCP establishment involves the subcellular polarization of core PCP components. It has been suggested that Wnt gradients could provide a global cue that coordinates local PCP with tissue axes. Here, we dissect the role of Wnt ligands in the orientation of hairs of Drosophila wings, an established system for the study of PCP. We found that PCP was normal in quintuple mutant wings that rely solely on the membrane-tethered Wingless for Wnt signaling, suggesting that a Wnt gradient is not required. We then used a nanobody-based approach to trap Wntless in the endoplasmic reticulum, and hence prevent all Wnt secretion, specifically during the period of PCP establishment. PCP was still established. We conclude that, even though Wnt ligands could contribute to PCP, they are not essential, and another global cue must exist for tissue-wide polarization.
Collapse
Affiliation(s)
| | - Aude Maugarny-Calès
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne University, CNRS UMR 3215, INSERM U934, 75005 Paris, France
| | - Stéphane Pelletier
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne University, CNRS UMR 3215, INSERM U934, 75005 Paris, France
| | | | - Yohanns Bellaiche
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne University, CNRS UMR 3215, INSERM U934, 75005 Paris, France
| | | | | |
Collapse
|
29
|
Ma SKY, Chan ASF, Rubab A, Chan WCW, Chan D. Extracellular Matrix and Cellular Plasticity in Musculoskeletal Development. Front Cell Dev Biol 2020; 8:781. [PMID: 32984311 PMCID: PMC7477050 DOI: 10.3389/fcell.2020.00781] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Cellular plasticity refers to the ability of cell fates to be reprogrammed given the proper signals, allowing for dedifferentiation or transdifferentiation into different cell fates. In vitro, this can be induced through direct activation of gene expression, however this process does not naturally occur in vivo. Instead, the microenvironment consisting of the extracellular matrix (ECM) and signaling factors, directs the signals presented to cells. Often the ECM is involved in regulating both biochemical and mechanical signals. In stem cell populations, this niche is necessary for maintenance and proper function of the stem cell pool. However, recent studies have demonstrated that differentiated or lineage restricted cells can exit their current state and transform into another state under different situations during development and regeneration. This may be achieved through (1) cells responding to a changing niche; (2) cells migrating and encountering a new niche; and (3) formation of a transitional niche followed by restoration of the homeostatic niche to sequentially guide cells along the regenerative process. This review focuses on examples in musculoskeletal biology, with the concept of ECM regulating cells and stem cells in development and regeneration, extending beyond the conventional concept of small population of progenitor cells, but under the right circumstances even “lineage-restricted” or differentiated cells can be reprogrammed to enter into a different fate.
Collapse
Affiliation(s)
- Sophia Ka Yan Ma
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | | | - Aqsa Rubab
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Wilson Cheuk Wing Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,Department of Orthopedics Surgery and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| |
Collapse
|
30
|
Evgrafov OV, Armoskus C, Wrobel BB, Spitsyna VN, Souaiaia T, Herstein JS, Walker CP, Nguyen JD, Camarena A, Weitz JR, Kim JMH, Lopez Duarte E, Wang K, Simpson GM, Sobell JL, Medeiros H, Pato MT, Pato CN, Knowles JA. Gene Expression in Patient-Derived Neural Progenitors Implicates WNT5A Signaling in the Etiology of Schizophrenia. Biol Psychiatry 2020; 88:236-247. [PMID: 32143829 PMCID: PMC10947993 DOI: 10.1016/j.biopsych.2020.01.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 01/03/2020] [Accepted: 01/06/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Genome-wide association studies of schizophrenia have demonstrated that variations in noncoding regions are responsible for most of the common variation heritability of the disease. It is hypothesized that these risk variants alter gene expression. Therefore, studying alterations in gene expression in schizophrenia may provide a direct approach to understanding the etiology of the disease. In this study we use cultured neural progenitor cells derived from olfactory neuroepithelium (CNON cells) as a genetically unaltered cellular model to elucidate the neurodevelopmental aspects of schizophrenia. METHODS We performed a gene expression study using RNA sequencing of CNON cells from 111 control subjects and 144 individuals with schizophrenia. Differentially expressed genes were identified with DESeq2 software, using covariates to correct for sex, age, library batches, and 1 surrogate variable component. RESULTS A total of 80 genes were differentially expressed (false discovery rate < 10%), showing enrichment in cell migration, cell adhesion, developmental process, synapse assembly, cell proliferation, and related Gene Ontology categories. Cadherin and Wnt signaling pathways were positive in overrepresentation test, and, in addition, many genes were specifically involved in WNT5A signaling. The differentially expressed genes were modestly, but significantly, enriched in the genes overlapping single nucleotide polymorphisms with genome-wide significant association from the Psychiatric Genomics Consortium genome-wide association study of schizophrenia. We also found substantial overlap with genes associated with other psychiatric disorders or brain development, enrichment in the same Gene Ontology categories as genes with mutations de novo in schizophrenia, and studies of induced pluripotent stem cell-derived neural progenitor cells. CONCLUSIONS CNON cells are a good model of the neurodevelopmental aspects of schizophrenia and can be used to elucidate the etiology of the disorder.
Collapse
Affiliation(s)
- Oleg V Evgrafov
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York.
| | - Chris Armoskus
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Bozena B Wrobel
- Caruso Department of Otolaryngology, Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Valeria N Spitsyna
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Tade Souaiaia
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - Jennifer S Herstein
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Christopher P Walker
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Joseph D Nguyen
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Adrian Camarena
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jonathan R Weitz
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jae Mun Hugo Kim
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Edder Lopez Duarte
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - George M Simpson
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Janet L Sobell
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Helena Medeiros
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - Michele T Pato
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - Carlos N Pato
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - James A Knowles
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York
| |
Collapse
|
31
|
Haumann I, Sturm MA, Anstötz M, Rune GM. GPER1 Signaling Initiates Migration of Female V-SVZ-Derived Cells. iScience 2020; 23:101077. [PMID: 32361597 PMCID: PMC7200306 DOI: 10.1016/j.isci.2020.101077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/01/2019] [Accepted: 04/14/2020] [Indexed: 12/30/2022] Open
Abstract
In the rodent ventricular-subventricular zone (V-SVZ) neurons are generated throughout life. They migrate along the rostral migratory stream (RMS) into the olfactory bulb before their final differentiation into interneurons and integration into local circuits. Estrogen receptors (ERs) are steroid hormone receptors with important functions in neurogenesis and synaptic plasticity. In this study, we show that the ER GPER1 is expressed in subsets of cells within the V-SVZ of female animals and provide evidence for a potential local estrogen source from aromatase-positive astrocytes surrounding the RMS. Blocking of GPER1 in Matrigel cultures of female animals significantly impairs migration of V-SVZ-derived cells. This outgrowth is accompanied by regulation of phosphorylation of the actin-binding protein cofilin by GPER1 signaling including an involvement of the p21-Ras pathway. Our results point to a prominent role of GPER1 in the initiation of neuronal migration from the V-SVZ to the olfactory bulb. GPER1 is expressed within all cell types of the stem cell lineage in the V-SVZ Blocking of GPER1 leads to a decrease in migration of V-SVZ-derived neuroblasts GPER1 signaling in V-SVZ Matrigel cultures involves Ras-induced p21 Blocking of GPER1 signaling leads to an increase in the ratio of p-cofilin/cofilin
Collapse
Affiliation(s)
- Iris Haumann
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Muriel Anne Sturm
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Max Anstötz
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| |
Collapse
|
32
|
Leroyer AS, Blin MG, Bachelier R, Bardin N, Blot-Chabaud M, Dignat-George F. CD146 (Cluster of Differentiation 146). Arterioscler Thromb Vasc Biol 2020; 39:1026-1033. [PMID: 31070478 DOI: 10.1161/atvbaha.119.312653] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CD146 (cluster of differentiation 146) is an adhesion molecule that is expressed by different cells constituting vessels, particularly endothelial cells. The last 30 years of research in this field have shown that CD146 plays a key role in the control of several vessel functions. Three forms of CD146 have been described, including 2 transmembrane isoforms and a soluble protein that is detectable in the plasma. These CD146 forms mediate pleiotropic functions through homophilic and heterophilic interactions with proteins present on surrounding partners. Several studies used neutralizing antibodies, siRNA, or genetically modified mice to demonstrate the involvement of CD146 in the regulation of angiogenesis, vascular permeability, and leukocyte transmigration. In this review, we will focus on the current knowledge of the roles of CD146 in vascular homeostasis and diseases associated with endothelial dysfunction.
Collapse
Affiliation(s)
- Aurélie S Leroyer
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Muriel G Blin
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Richard Bachelier
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Nathalie Bardin
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.).,Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, France (N.B., F.D.-G.)
| | - Marcel Blot-Chabaud
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Françoise Dignat-George
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.).,Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, France (N.B., F.D.-G.)
| |
Collapse
|
33
|
Jati S, Sarraf TR, Naskar D, Sen M. Wnt Signaling: Pathogen Incursion and Immune Defense. Front Immunol 2019; 10:2551. [PMID: 31736969 PMCID: PMC6828841 DOI: 10.3389/fimmu.2019.02551] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/15/2019] [Indexed: 12/15/2022] Open
Abstract
Wnt ligands interact with the transmembrane cell surface receptors Frizzled and ROR/RYK to initiate complex signaling cascades that are crucial for cell physiology and the proper functioning of the immune system. Wnt signaling is instrumental in maintaining immune surveillance and during infections by pathogenic microbes helps mount host resistance to infection. Some pathogens, however, utilize Wnt signaling to build a niche for their survival. The goal of this review is to summarize current and developing concepts about the tug of war between Wnt signaling and pathogens for deployment of host resources, focusing mostly on macrophages and cytoskeletal actin dynamics. An additional objective is to outline the interrelation between Wnt signaling and the host microbiota, which is vital for immune defense, discussing in the same perspective, how Wnt signaling could be differentiating pathogen from non-pathogen.
Collapse
Affiliation(s)
- Suborno Jati
- Division of Cancer Biology and Inflammatory Disorder, Indian Institute of Chemical Biology, Kolkata, India
| | - Tresa Rani Sarraf
- Division of Cancer Biology and Inflammatory Disorder, Indian Institute of Chemical Biology, Kolkata, India
| | - Debdut Naskar
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Kolkata, India
| | - Malini Sen
- Division of Cancer Biology and Inflammatory Disorder, Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
34
|
Rogan MR, Patterson LL, Wang JY, McBride JW. Bacterial Manipulation of Wnt Signaling: A Host-Pathogen Tug-of-Wnt. Front Immunol 2019; 10:2390. [PMID: 31681283 PMCID: PMC6811524 DOI: 10.3389/fimmu.2019.02390] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/23/2019] [Indexed: 12/27/2022] Open
Abstract
The host-pathogen interface is a crucial battleground during bacterial infection in which host defenses are met with an array of bacterial counter-mechanisms whereby the invader aims to make the host environment more favorable to survival and dissemination. Interestingly, the eukaryotic Wnt signaling pathway has emerged as a key player in the host and pathogen tug-of-war. Although studied for decades as a regulator of embryogenesis, stem cell maintenance, bone formation, and organogenesis, Wnt signaling has recently been shown to control processes related to bacterial infection in the human host. Wnt signaling pathways contribute to cell cycle control, cytoskeleton reorganization during phagocytosis and cell migration, autophagy, apoptosis, and a number of inflammation-related events. Unsurprisingly, bacterial pathogens have evolved strategies to manipulate these Wnt-associated processes in order to enhance infection and survival within the human host. In this review, we examine the different ways human bacterial pathogens with distinct host cell tropisms and lifestyles exploit Wnt signaling for infection and address the potential of harnessing Wnt-related mechanisms to combat infectious disease.
Collapse
Affiliation(s)
- Madison R. Rogan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - LaNisha L. Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jennifer Y. Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
35
|
Heikenwalder M, Lorentzen A. The role of polarisation of circulating tumour cells in cancer metastasis. Cell Mol Life Sci 2019; 76:3765-3781. [PMID: 31218452 PMCID: PMC6744547 DOI: 10.1007/s00018-019-03169-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/23/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
Metastasis is the spread of cancer cells from a primary tumour to a distant site of the body. Metastasising tumour cells have to survive and readjust to different environments, such as heterogeneous solid tissues and liquid phase in lymph- or blood circulation, which they achieve through a high degree of plasticity that renders them adaptable to varying conditions. One defining characteristic of the metastatic process is the transition of tumour cells between different polarised phenotypes, ranging from differentiated epithelial polarity to migratory front-rear polarity. Here, we review the polarisation types adopted by tumour cells during the metastatic process and describe the recently discovered single-cell polarity in liquid phase observed in circulating tumour cells. We propose that single-cell polarity constitutes a mode of polarisation of the cell cortex that is uncoupled from the intracellular polarisation machinery, which distinguishes single-cell polarity from other types of polarity identified so far. We discuss how single-cell polarity can contribute to tumour metastasis and the therapeutic potential of this new discovery.
Collapse
Affiliation(s)
- Mathias Heikenwalder
- Divison of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
| | - Anna Lorentzen
- Department of Molecular Biology and Genetics, Aarhus University, 8000, Aarhus, Denmark.
| |
Collapse
|
36
|
Wu X, Yan T, Hao L, Zhu Y. Wnt5a induces ROR1 and ROR2 to activate RhoA in esophageal squamous cell carcinoma cells. Cancer Manag Res 2019; 11:2803-2815. [PMID: 31114334 PMCID: PMC6497886 DOI: 10.2147/cmar.s190999] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/27/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Wnt5a is a nontransforming Wnt family member and identified as an oncogenic role on cell motility of breast cancer and glioblastoma. However, Wnt5a signaling in esophageal squamous cell carcinoma (ESCC) progression remains poorly defined. Materials and methods: Immunohistochemistry assays were used to measure the Wnt5a expression in ESCC sections. We evaluated the role of receptor tyrosine kinase-like orphan receptor (ROR)1/2 and RhoA on the invasion of ESCC cells by using cell invasion assay, immunoprecipitation, immunofluorescence, and Rho activation assay. Results: Wnt5a was highly expressed in invasive ESCC tissues compared with that in noninvasive and nonmalignant tissues. In vitro assay showed that sfrp2 (Wnt5a antagonist) largely blocked the invasion but not the colony formation of KYSE410 and KYSE520 ESCC cells. Anti-ROR1 mAb and ROR2-shRNA markedly inhibited the disheveled-associated activator of morphogenesis 1 (DAAM1) activity, RhoA activity, microfilament formation and the invasion of ESCC cells. Fluorescent phalloidin staining experiment showed ROR1/ROR2, receptors of Wnt5a signaling, and regulated the reassembly of actin filaments in ESCC cells. Further experiments showed that ROR1 was strongly associated with ROR2 in KYSE410 cells. The activation of RhoA, not Rac1 or Rac2, was involved in ROR1/ROR2 signaling pathway. By using DAAM1 shRNA, we found that RhoA was downstream of DAAM1, which could be rescued by the overexpression of wild-type DAAM1. This could be further proved by a RhoA inhibitor CCG-1423 which could inhibit the invasion of ESCC cells but not DAAM1 activity. Conclusions: Wnt5a promotes ESCC cell invasion via ROR1 and ROR2 receptors and DAAM1/RhoA signaling pathway.
Collapse
Affiliation(s)
- Xuping Wu
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, People's Republic of China
| | - Ting Yan
- Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Leiyu Hao
- Department of Physiology, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing 211166, People's Republic of China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, People's Republic of China
| |
Collapse
|
37
|
Hasan MK, Rassenti L, Widhopf GF, Yu J, Kipps TJ. Wnt5a causes ROR1 to complex and activate cortactin to enhance migration of chronic lymphocytic leukemia cells. Leukemia 2018; 33:653-661. [PMID: 30568170 PMCID: PMC6462876 DOI: 10.1038/s41375-018-0306-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 09/15/2018] [Accepted: 11/14/2018] [Indexed: 01/29/2023]
Abstract
Chronic lymphocytic leukemia cells (CLL) migrate between the blood and lymphoid tissues in response to chemokines. Such migration requires structured cytoskeletal-actin polymerization, which may involve the protein cortactin. We discovered that treatment of CLL cells with Wnt5a causes Receptor tyosin kinase-like orphan receptor 1 (ROR1) to bind cortactin, which undergoes tyrosine phosphorylation at Y421, recruits ARHGEF1, and activates RhoA, thereby enhancing leukemia-cell migration; such effects could be inhibited by cirmtuzumab, a humanized mAb specific for ROR1. We transfected the CLL-cell-line MEC1 with either full-length ROR1 or various mutant forms of ROR1 to examine the structural features required for binding cortactin. We found that the proline-rich domain (PRD) was necessary for ROR1 to recruit cortactin. We generated MEC1 cells that each expressed a mutant form of ROR1 with a single amino-acid substitution of alanine (A) for proline (P) in potential SH3-binding sites in the ROR1-PRD at positions 784, 808, 826, 841, or 850. In contrast to wild-type ROR1, or other ROR1P=>A mutants, ROR1P(841)A failed to complex with cortactin or ARHGEF1 in response to Wnt5a. Moreover, Wnt5a could not induce MEC1-ROR1P(841)A to phosphorylate cortactin or enhance CLL-cell F-actin polymerization. Taken together, these studies show that cortactin plays an important role in ROR1-dependent Wnt5a-enhanced CLL-cell migration.
Collapse
Affiliation(s)
- Md Kamrul Hasan
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Laura Rassenti
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - George F Widhopf
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Jian Yu
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Thomas J Kipps
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
38
|
Vasquez YM, Wang X, Wetendorf M, Franco HL, Mo Q, Wang T, Lanz RB, Young SL, Lessey BA, Spencer TE, Lydon JP, DeMayo FJ. FOXO1 regulates uterine epithelial integrity and progesterone receptor expression critical for embryo implantation. PLoS Genet 2018; 14:e1007787. [PMID: 30452456 PMCID: PMC6277115 DOI: 10.1371/journal.pgen.1007787] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 12/03/2018] [Accepted: 10/25/2018] [Indexed: 12/11/2022] Open
Abstract
Successful embryo implantation requires a receptive endometrium. Poor uterine receptivity can account for implantation failure in women who experience recurrent pregnancy loss or multiple rounds of unsuccessful in vitro fertilization cycles. Here, we demonstrate that the transcription factor Forkhead Box O1 (FOXO1) is a critical regulator of endometrial receptivity in vivo. Uterine ablation of Foxo1 using the progesterone receptor Cre (PgrCre) mouse model resulted in infertility due to altered epithelial cell polarity and apoptosis, preventing the embryo from penetrating the luminal epithelium. Analysis of the uterine transcriptome after Foxo1 ablation identified alterations in gene expression for transcripts involved in the activation of cell invasion, molecular transport, apoptosis, β-catenin (CTNNB1) signaling pathway, and an increase in PGR signaling. The increase of PGR signaling was due to PGR expression being retained in the uterine epithelium during the window of receptivity. Constitutive expression of epithelial PGR during this receptive period inhibited expression of FOXO1 in the nucleus of the uterine epithelium. The reciprocal expression of PGR and FOXO1 was conserved in human endometrial samples during the proliferative and secretory phase. This demonstrates that expression of FOXO1 and the loss of PGR during the window of receptivity are interrelated and critical for embryo implantation.
Collapse
Affiliation(s)
- Yasmin M. Vasquez
- Department of Molecular and Cellular Biology and Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Xiaoqiu Wang
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States of America
- Department of Animal Science, North Carolina State University, Raleigh, NC, United States of America
| | - Margeaux Wetendorf
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States of America
| | - Heather L. Franco
- Department of Molecular and Cellular Biology and Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Qianxing Mo
- Department of Medicine and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Tianyuan Wang
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States of America
| | - Rainer B. Lanz
- Department of Molecular and Cellular Biology and Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Steven L. Young
- Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Bruce A. Lessey
- Department of Obstetrics and Gynecology, University of South Carolina School of Medicine, Greenville, SC, United States of America
| | - Thomas E. Spencer
- Division of Animal Science, University of Missouri, Columbia, MO, United States of America
| | - John P. Lydon
- Department of Molecular and Cellular Biology and Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Francesco J. DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States of America
| |
Collapse
|
39
|
Sivakumar A, Mahadevan A, Lauer ME, Narvaez RJ, Ramesh S, Demler CM, Souchet NR, Hascall VC, Midura RJ, Garantziotis S, Frank DB, Kimata K, Kurpios NA. Midgut Laterality Is Driven by Hyaluronan on the Right. Dev Cell 2018; 46:533-551.e5. [PMID: 30174180 PMCID: PMC6207194 DOI: 10.1016/j.devcel.2018.08.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 06/01/2018] [Accepted: 08/01/2018] [Indexed: 11/24/2022]
Abstract
For many years, biologists have focused on the role of Pitx2, expressed on the left side of developing embryos, in governing organ laterality. Here, we identify a different pathway during left-right asymmetry initiated by the right side of the embryo. Surprisingly, this conserved mechanism is orchestrated by the extracellular glycosaminoglycan, hyaluronan (HA) and is independent of Pitx2 on the left. Whereas HA is normally synthesized bilaterally as a simple polysaccharide, we show that covalent modification of HA by the enzyme Tsg6 on the right triggers distinct cell behavior necessary to drive the conserved midgut rotation and to pattern gut vasculature. HA disruption in chicken and Tsg6-/- mice results in failure to initiate midgut rotation and perturbs vascular development predisposing to midgut volvulus. Our study leads us to revise the current symmetry-breaking paradigm in vertebrates and demonstrates how enzymatic modification of HA matrices can execute the blueprint of organ laterality.
Collapse
Affiliation(s)
- Aravind Sivakumar
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Aparna Mahadevan
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Mark E Lauer
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Ricky J Narvaez
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Siddesh Ramesh
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Cora M Demler
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Nathan R Souchet
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Vincent C Hascall
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Ron J Midura
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Stavros Garantziotis
- Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - David B Frank
- Division of Pediatric Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Koji Kimata
- Institute of Molecular Medical Sciences, Aichi Medical University, Nagakute, Aichi, Japan
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
40
|
Gao B, Ajima R, Yang W, Li C, Song H, Anderson MJ, Liu RR, Lewandoski MB, Yamaguchi TP, Yang Y. Coordinated directional outgrowth and pattern formation by integration of Wnt5a and Fgf signaling in planar cell polarity. Development 2018; 145:dev.163824. [PMID: 29615464 DOI: 10.1242/dev.163824] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/19/2018] [Indexed: 12/28/2022]
Abstract
Embryonic morphogenesis of a complex organism requires proper regulation of patterning and directional growth. Planar cell polarity (PCP) signaling is emerging as a crucial evolutionarily conserved mechanism whereby directional information is conveyed. PCP is thought to be established by global cues, and recent studies have revealed an instructive role of a Wnt signaling gradient in epithelial tissues of both invertebrates and vertebrates. However, it remains unclear whether Wnt/PCP signaling is regulated in a coordinated manner with embryonic patterning during morphogenesis. Here, in mouse developing limbs, we find that apical ectoderm ridge-derived Fgfs required for limb patterning regulate PCP along the proximal-distal axis in a Wnt5a-dependent manner. We demonstrate with genetic evidence that the Wnt5a gradient acts as a global cue that is instructive in establishing PCP in the limb mesenchyme, and that Wnt5a also plays a permissive role to allow Fgf signaling to orient PCP. Our results indicate that limb morphogenesis is regulated by coordination of directional growth and patterning through integration of Wnt5a and Fgf signaling.
Collapse
Affiliation(s)
- Bo Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China .,Developmental Genetics Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Rieko Ajima
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, NIH, Frederick, MD 21702, USA
| | - Wei Yang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Chunyu Li
- Developmental Genetics Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA.,Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Hai Song
- Developmental Genetics Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Matthew J Anderson
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, NIH, Frederick, MD 21702, USA
| | - Robert R Liu
- Developmental Genetics Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Mark B Lewandoski
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, NIH, Frederick, MD 21702, USA
| | - Terry P Yamaguchi
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, NIH, Frederick, MD 21702, USA
| | - Yingzi Yang
- Developmental Genetics Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA .,Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
41
|
Sadeghi RS, Kulej K, Kathayat RS, Garcia BA, Dickinson BC, Brady DC, Witze ES. Wnt5a signaling induced phosphorylation increases APT1 activity and promotes melanoma metastatic behavior. eLife 2018; 7:34362. [PMID: 29648538 PMCID: PMC5919757 DOI: 10.7554/elife.34362] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/11/2018] [Indexed: 11/13/2022] Open
Abstract
Wnt5a has been implicated in melanoma progression and metastasis, although the exact downstream signaling events that contribute to melanoma metastasis are poorly understood. Wnt5a signaling results in acyl protein thioesterase 1 (APT1) mediated depalmitoylation of pro-metastatic cell adhesion molecules CD44 and MCAM, resulting in increased melanoma invasion. The mechanistic details that underlie Wnt5a-mediated regulation of APT1 activity and cellular function remain unknown. Here, we show Wnt5a signaling regulates APT1 activity through induction of APT1 phosphorylation and we further investigate the functional role of APT1 phosphorylation on its depalmitoylating activity. We found phosphorylation increased APT1 depalmitoylating activity and reduced APT1 dimerization. We further determined APT1 phosphorylation increases melanoma invasion in vitro, and also correlated with increased tumor grade and metastasis. Our results further establish APT1 as an important regulator of melanoma invasion and metastatic behavior. Inhibition of APT1 may represent a novel way to treat Wnt5a driven cancers.
Collapse
Affiliation(s)
- Rochelle Shirin Sadeghi
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Katarzyna Kulej
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | | | - Benjamin A Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Bryan C Dickinson
- Department of Chemistry, University of Chicago, Chicago, United States
| | - Donita C Brady
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Eric S Witze
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
42
|
Jati S, Kundu S, Chakraborty A, Mahata SK, Nizet V, Sen M. Wnt5A Signaling Promotes Defense Against Bacterial Pathogens by Activating a Host Autophagy Circuit. Front Immunol 2018; 9:679. [PMID: 29686674 PMCID: PMC5900007 DOI: 10.3389/fimmu.2018.00679] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/20/2018] [Indexed: 12/14/2022] Open
Abstract
Bacterial pathogens are associated with severe infections (e.g., sepsis) and exacerbation of debilitating conditions such as chronic obstructive pulmonary disease (COPD). The interactions of bacterial pathogens with macrophages, a key component of innate immunity and host defense, are not clearly understood and continue to be intensively studied. Having previously demonstrated a role of Wnt5A signaling in phagocytosis, we proceeded to decipher the connection of Wnt5A signaling with infection by pathogenic bacteria, namely Pseudomonas aeruginosa (PA) and Streptococcus pneumoniae (SP), which are related with the progression of COPD and sepsis. We found that during the initial hours of infection with PA and SP, there is decrease in the steady state levels of the Wnt5A protein in macrophages. Suppression of Wnt5A signaling, moreover, impairs macrophage clearance of the bacterial infection both in vitro and in vivo. Activation of Wnt5A signaling, on the other hand, enhances clearance of the infection. Macrophage-mediated containment of bacterial infection in our study is dependant on Wnt5A-induced Rac1/Disheveled activation and cytochalasin D inhibitable actin assembly, which is associated with ULK1 kinase activity and LC3BII accumulation. Our experimental findings are consistent with Wnt5A signaling-dependent induction of autophagic killing (xenophagy) of PA and SP, as further substantiated by transmission electron microscopy. Overall, our study unveils the prevalence of a Wnt5A-Rac1-Disheveled-mediated actin-associated autophagy circuit as an important component of innate immunity in host macrophages that may turn out crucial for restricting infection by leading bacterial pathogens.
Collapse
Affiliation(s)
- Suborno Jati
- Division of Cancer Biology and Inflammatory Disorder, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Suman Kundu
- Division of Cancer Biology and Inflammatory Disorder, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Arijit Chakraborty
- Division of Cancer Biology and Inflammatory Disorder, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sushil Kumar Mahata
- Department of Medicine, VA San Diego Healthcare System and University of California, San Diego, La Jolla, CA, United States
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Malini Sen
- Division of Cancer Biology and Inflammatory Disorder, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
43
|
Sarin S, Zuniga-Sanchez E, Kurmangaliyev YZ, Cousins H, Patel M, Hernandez J, Zhang KX, Samuel MA, Morey M, Sanes JR, Zipursky SL. Role for Wnt Signaling in Retinal Neuropil Development: Analysis via RNA-Seq and In Vivo Somatic CRISPR Mutagenesis. Neuron 2018; 98:109-126.e8. [PMID: 29576390 DOI: 10.1016/j.neuron.2018.03.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 01/16/2018] [Accepted: 03/02/2018] [Indexed: 12/22/2022]
Abstract
Screens for genes that orchestrate neural circuit formation in mammals have been hindered by practical constraints of germline mutagenesis. To overcome these limitations, we combined RNA-seq with somatic CRISPR mutagenesis to study synapse development in the mouse retina. Here synapses occur between cellular layers, forming two multilayered neuropils. The outer neuropil, the outer plexiform layer (OPL), contains synapses made by rod and cone photoreceptor axons on rod and cone bipolar dendrites, respectively. We used RNA-seq to identify selectively expressed genes encoding cell surface and secreted proteins and CRISPR-Cas9 electroporation with cell-specific promoters to assess their roles in OPL development. Among the genes identified in this way are Wnt5a and Wnt5b. They are produced by rod bipolars and activate a non-canonical signaling pathway in rods to regulate early OPL patterning. The approach we use here can be applied to other parts of the brain.
Collapse
Affiliation(s)
- Sumeet Sarin
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Elizabeth Zuniga-Sanchez
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yerbol Z Kurmangaliyev
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Henry Cousins
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Mili Patel
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Jeanette Hernandez
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kelvin X Zhang
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Melanie A Samuel
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Marta Morey
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA.
| | - S Lawrence Zipursky
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
44
|
Single cell polarity in liquid phase facilitates tumour metastasis. Nat Commun 2018; 9:887. [PMID: 29491397 PMCID: PMC5830403 DOI: 10.1038/s41467-018-03139-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/19/2018] [Indexed: 01/19/2023] Open
Abstract
Dynamic polarisation of tumour cells is essential for metastasis. While the role of polarisation during dedifferentiation and migration is well established, polarisation of metastasising tumour cells during phases of detachment has not been investigated. Here we identify and characterise a type of polarisation maintained by single cells in liquid phase termed single-cell (sc) polarity and investigate its role during metastasis. We demonstrate that sc polarity is an inherent feature of cells from different tumour entities that is observed in circulating tumour cells in patients. Functionally, we propose that the sc pole is directly involved in early attachment, thereby affecting adhesion, transmigration and metastasis. In vivo, the metastatic capacity of cell lines correlates with the extent of sc polarisation. By manipulating sc polarity regulators and by generic depolarisation, we show that sc polarity prior to migration affects transmigration and metastasis in vitro and in vivo. Polarisation of metastasising cancer cells in circulation has not been investigated before. Here the authors identify single cell polarity as a distinct polarisation state of single cells in liquid phase, and show that perturbing single cell polarity affects attachment, adhesion, transmigration and metastasis in vitro and in vivo.
Collapse
|
45
|
Brown S, Matta A, Erwin M, Roberts S, Gruber HE, Hanley EN, Little CB, Melrose J. Cell Clusters Are Indicative of Stem Cell Activity in the Degenerate Intervertebral Disc: Can Their Properties Be Manipulated to Improve Intrinsic Repair of the Disc? Stem Cells Dev 2018; 27:147-165. [DOI: 10.1089/scd.2017.0213] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Sharon Brown
- Spinal Studies and ISTM (Keele University), Robert Jones and Agnes Hunt Orthopaedic Hospital, NHS Foundation Trust, Oswestry, United Kingdom
| | - Ajay Matta
- Krembil Research Institute, Toronto, Canada
| | - Mark Erwin
- Krembil Research Institute, Toronto, Canada
| | - Sally Roberts
- Spinal Studies and ISTM (Keele University), Robert Jones and Agnes Hunt Orthopaedic Hospital, NHS Foundation Trust, Oswestry, United Kingdom
| | - Helen E. Gruber
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Carolina
| | - Edward N. Hanley
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Carolina
| | - Christopher B. Little
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, The Royal North Shore Hospital, St. Leonards, NSW, Australia
- Sydney Medical School, Northern, The University of Sydney. Royal North Shore Hospital, St. Leonards, Australia
| | - James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, The Royal North Shore Hospital, St. Leonards, NSW, Australia
- Sydney Medical School, Northern, The University of Sydney. Royal North Shore Hospital, St. Leonards, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| |
Collapse
|
46
|
Demonstration of a WNT5A-IL-6 positive feedback loop in melanoma cells: Dual interference of this loop more effectively impairs melanoma cell invasion. Oncotarget 2018; 7:37790-37802. [PMID: 27191257 PMCID: PMC5122349 DOI: 10.18632/oncotarget.9332] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 04/28/2016] [Indexed: 01/07/2023] Open
Abstract
Increased expression and signalling of WNT5A and interleukin-6 (IL-6) have both been shown to promote melanoma progression. Here, we investigated the proposed existence of a WNT5A-IL-6 positive feedback loop that drives melanoma migration and invasion. First, the HOPP algorithm revealed that the invasive phenotype of cultured melanoma cells was significantly correlated with increased expression of WNT5A or IL-6. In three invasive melanoma cell lines, endogenous WNT5A protein expression was related to IL-6 protein secretion. Knockdown with anti-IL-6 siRNAs or treating WM852 melanoma cells with a neutralising anti-IL-6 antibody reduced WNT5A protein expression. Conversely, the silencing of WNT5A expression by WNT5A siRNAs or treating WM852 melanoma cells with Box5 (a WNT5A antagonist) significantly reduced IL-6 secretion. Interestingly, these effects occurred at the protein level but not at the transcriptional levels. Functionally, we demonstrated that combined siRNA knockdown of WNT5A and IL-6 expression or the simultaneous inhibition of WNT5A and IL-6 signalling inhibited melanoma cell invasion more effectively than suppressing each factor individually. Together, our results demonstrate that WNT5A and IL-6 are connected through a positive feedback loop in melanoma cells and that the combined targeting of both molecules could serve as an effective therapeutic means to reduce melanoma metastasis.
Collapse
|
47
|
Reimer M, Denby E, Zustiak SP, Schober JM. Ras GAP-related and C-terminal domain-dependent localization and tumorigenic activities of IQGAP1 in melanoma cells. PLoS One 2017; 12:e0189589. [PMID: 29240845 PMCID: PMC5730206 DOI: 10.1371/journal.pone.0189589] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/29/2017] [Indexed: 12/19/2022] Open
Abstract
IQGAP1 interacts with a number of binding partners through a calponin homology domain (CHD), a WW motif, IQ repeats, a Ras GAP-related domain (GRD), and a conserved C-terminal (CT) domain. Among various biological and cellular functions, IQGAP1 is known to play a role in actin cytoskeleton dynamics during membrane ruffling and lamellipodium protrusion. In addition, phosphorylation near the CT domain is thought to control IQGAP1 activity through regulation of intramolecular interaction. In a previous study, we discovered that IQGAP1 preferentially localizes to retracting areas in B16F10 mouse melanoma cells, not areas of membrane ruffling and lamellipodium protrusion. Nothing is known of the domains needed for retraction localization and very little is known of IQGAP1 function in the actin cytoskeleton of melanoma cells. Thus, we examined localization of IQGAP1 mutants to retracting areas, and characterized knock down phenotypes on tissue culture plastic and physiologic-stiffness hydrogels. Localization of IQGAP1 mutants (S1441E/S1443D, S1441A/S1443A, ΔCHD, ΔGRD or ΔCT) to retracting and protruding cell edges were measured. In retracting areas there was a decrease in S1441A/S1443A, ΔGRD and ΔCT localization, a minor decrease in ΔCHD localization, and normal localization of the S1441E/S1443D mutant. In areas of cell protrusion just behind the lamellipodium leading edge, we surprisingly observed both ΔGRD and ΔCT localization, and increased number of microtubules. IQGAP1 knock down caused loss of cell polarity on laminin-coated glass, decreased proliferation on tissue culture polystyrene, and abnormal spheroid growth on laminin-coated hydrogels. We propose that the GRD and CT domains regulate IQGAP1 localization to retracting actin networks to promote a tumorigenic role in melanoma cells.
Collapse
Affiliation(s)
- Michael Reimer
- Department of Pharmaceutical Sciences, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America
| | - Elisabeth Denby
- Department of Pharmaceutical Sciences, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America
| | - Silviya P. Zustiak
- Department of Biomedical Engineering, Saint Louis University, Saint Louis, Missouri, United States of America
| | - Joseph M. Schober
- Department of Pharmaceutical Sciences, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America
- * E-mail:
| |
Collapse
|
48
|
Liu G, Yan T, Li X, Sun J, Zhang B, Wang H, Zhu Y. Daam1 activates RhoA to regulate Wnt5a‑induced glioblastoma cell invasion. Oncol Rep 2017; 39:465-472. [PMID: 29207169 PMCID: PMC5783613 DOI: 10.3892/or.2017.6124] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 09/27/2017] [Indexed: 02/04/2023] Open
Abstract
The signaling pathway of dishevelled-associated activator of morphogenesis 1 (Daam1) triggered by Wnt5a drives cell movement and migration during breast cancer metastasis. However, Wnt5a signaling in glioblastoma progression remains poorly defined. Wnt5a expression and activations of RhoA, Cdc42, and Rac1 were detected in human glioblastoma tissues by using ELISA assays and small G-protein activation assays, respectively. The cell invasion rate and Daam1 activation of glioblastoma U251 and T98MG cells were determined by cell invasion assays and pull-down assays, respectively. According to our experiments, Wnt5a expression and RhoA activation were upregulated in invasive glioblastoma tissues, with a significant positive correlation between them. Wnt5a activated Daam1 and RhoA, and subsequently promoted the invasion of glioblastoma U251 and T98MG cells. This process was abolished by secreted frizzled-related protein 2 (sFRP2), an antagonist that directly binds to Wnt5a. Specific small interfering RNA (siRNA) targeting Daam1 markedly inhibited Wnt5a-induced RhoA activation, stress fiber formation and glioblastoma cell invasion. CCG-1423, a RhoA inhibitor, decreased Wnt5a-induced stress fiber formation and glioblastoma cell invasion. Finally, siRNA targeting Daam1 or CCG-1423 treatment did not alter the cell proliferation of glioblastoma U251 and T98MG cells. We thus concluded that Wnt5a promoted glioblastoma cell invasion via Daam1/RhoA signaling pathway.
Collapse
Affiliation(s)
- Guiyang Liu
- Department of Neurosurgery, Jinan Fourth People's Hospital, Jinan, Shandong 250031, P.R. China
| | - Ting Yan
- Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drugs of Jiangsu Province, School of Public Health, Nanjing, Jiangsu 211166, P.R. China
| | - Xiaorong Li
- Department of Neurosurgery, Jinan Fourth People's Hospital, Jinan, Shandong 250031, P.R. China
| | - Jianhui Sun
- Department of Neurosurgery, Jinan Fourth People's Hospital, Jinan, Shandong 250031, P.R. China
| | - Bo Zhang
- Department of Neurosurgery, Jinan Fourth People's Hospital, Jinan, Shandong 250031, P.R. China
| | - Hongjie Wang
- Department of Neurosurgery, Jinan Fourth People's Hospital, Jinan, Shandong 250031, P.R. China
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
49
|
Moreno-Fortuny A, Bragg L, Cossu G, Roostalu U. MCAM contributes to the establishment of cell autonomous polarity in myogenic and chondrogenic differentiation. Biol Open 2017; 6:1592-1601. [PMID: 28923978 PMCID: PMC5703611 DOI: 10.1242/bio.027771] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell polarity has a fundamental role in shaping the morphology of cells and growing tissues. Polarity is commonly thought to be established in response to extracellular signals. Here we used a minimal in vitro assay that enabled us to monitor the determination of cell polarity in myogenic and chondrogenic differentiation in the absence of external signalling gradients. We demonstrate that the initiation of cell polarity is regulated by melanoma cell adhesion molecule (MCAM). We found highly polarized localization of MCAM, Moesin (MSN), Scribble (SCRIB) and Van-Gogh-like 2 (VANGL2) at the distal end of elongating myotubes. Knockout of MCAM or elimination of its endocytosis motif does not impair the initiation of myogenesis or myoblast fusion, but prevents myotube elongation. MSN, SCRIB and VANGL2 remain uniformly distributed in MCAM knockout cells. We show that MCAM is also required at early stages of chondrogenic differentiation. In both myogenic and chondrogenic differentiation MCAM knockout leads to transcriptional downregulation of Scrib and enhanced MAP kinase activity. Our data demonstrates the importance of cell autonomous polarity in differentiation. Summary: CD146/MCAM regulates cell autonomous polarization and asymmetric localization of Scribble, Van-Gogh-like 2 and Moesin, which is required in skeletal muscle myotube elongation and chondrocyte differentiation.
Collapse
Affiliation(s)
- Artal Moreno-Fortuny
- Manchester Academic Health Science Centre, Division of Extracellular Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Laricia Bragg
- Manchester Academic Health Science Centre, Division of Extracellular Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Giulio Cossu
- Manchester Academic Health Science Centre, Division of Extracellular Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Urmas Roostalu
- Manchester Academic Health Science Centre, Division of Extracellular Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| |
Collapse
|
50
|
Susman MW, Karuna EP, Kunz RC, Gujral TS, Cantú AV, Choi SS, Jong BY, Okada K, Scales MK, Hum J, Hu LS, Kirschner MW, Nishinakamura R, Yamada S, Laird DJ, Jao LE, Gygi SP, Greenberg ME, Ho HYH. Kinesin superfamily protein Kif26b links Wnt5a-Ror signaling to the control of cell and tissue behaviors in vertebrates. eLife 2017; 6:e26509. [PMID: 28885975 PMCID: PMC5590807 DOI: 10.7554/elife.26509] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022] Open
Abstract
Wnt5a-Ror signaling constitutes a developmental pathway crucial for embryonic tissue morphogenesis, reproduction and adult tissue regeneration, yet the molecular mechanisms by which the Wnt5a-Ror pathway mediates these processes are largely unknown. Using a proteomic screen, we identify the kinesin superfamily protein Kif26b as a downstream target of the Wnt5a-Ror pathway. Wnt5a-Ror, through a process independent of the canonical Wnt/β-catenin-dependent pathway, regulates the cellular stability of Kif26b by inducing its degradation via the ubiquitin-proteasome system. Through this mechanism, Kif26b modulates the migratory behavior of cultured mesenchymal cells in a Wnt5a-dependent manner. Genetic perturbation of Kif26b function in vivo caused embryonic axis malformations and depletion of primordial germ cells in the developing gonad, two phenotypes characteristic of disrupted Wnt5a-Ror signaling. These findings indicate that Kif26b links Wnt5a-Ror signaling to the control of morphogenetic cell and tissue behaviors in vertebrates and reveal a new role for regulated proteolysis in noncanonical Wnt5a-Ror signal transduction.
Collapse
Affiliation(s)
- Michael W Susman
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| | - Edith P Karuna
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Ryan C Kunz
- Department of Cell BiologyHarvard Medical SchoolBostonUnited States
| | - Taranjit S Gujral
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
- Division of Human BiologyFred Hutchinson Cancer Research CenterSeattleUnited States
| | - Andrea V Cantú
- Department of Obstetrics, Gynecology and Reproductive SciencesCenter for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of CaliforniaSan FranciscoUnited States
| | - Shannon S Choi
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Brigette Y Jong
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Kyoko Okada
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Michael K Scales
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Jennie Hum
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Linda S Hu
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| | - Marc W Kirschner
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
| | - Ryuichi Nishinakamura
- Department of Kidney DevelopmentInstitute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Soichiro Yamada
- Department of Biomedical EngineeringUniversity of CaliforniaDavisUnited States
| | - Diana J Laird
- Department of Obstetrics, Gynecology and Reproductive SciencesCenter for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of CaliforniaSan FranciscoUnited States
| | - Li-En Jao
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Steven P Gygi
- Department of Cell BiologyHarvard Medical SchoolBostonUnited States
| | | | - Hsin-Yi Henry Ho
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| |
Collapse
|